Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
BMC Genomics ; 25(1): 353, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594632

ABSTRACT

Mosquitoes are prolific vectors of human pathogens, therefore a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster, is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae (s.l.) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti, however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.


Subject(s)
Aedes , Bacterial Infections , Mycoses , Animals , Humans , Drosophila melanogaster , Mosquito Vectors/genetics , Aedes/genetics , Aedes/microbiology , Bacteria , Fungi/genetics
2.
Philos Trans R Soc Lond B Biol Sci ; 379(1901): 20230063, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38497256

ABSTRACT

Discussions of host-microbe interactions in mosquito vectors are frequently dominated by a focus on the human pathogens they transmit (e.g. Plasmodium parasites and arboviruses). Underlying the interactions between a vector and its transmissible pathogens, however, is the physiology of an insect living and interacting with a world of bacteria and fungi including commensals, mutualists and primary and opportunistic pathogens. Here we review what is known about the bacteria and fungi associated with mosquitoes, with an emphasis on the members of the Aedes genus. We explore the reciprocal effects of microbe on mosquito, and mosquito on microbe. We analyse the roles of bacterial and fungal symbionts in mosquito development, their effects on vector competence, and their potential uses as biocontrol agents and vectors for paratransgenesis. We explore the compartments of the mosquito gut, uncovering the regionalization of immune effectors and modulators, which create the zones of resistance and immune tolerance with which the mosquito host controls and corrals its microbial symbionts. We examine the anatomical patterning of basally expressed antimicrobial peptides. Finally, we review the relationships between inducible antimicrobial peptides and canonical immune signalling pathways, comparing and contrasting current knowledge on each pathway in mosquitoes to the model insect Drosophila melanogaster. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.


Subject(s)
Aedes , Microbiota , Animals , Humans , Drosophila melanogaster , Bacteria , Immunity, Innate , Antimicrobial Peptides
3.
bioRxiv ; 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37546902

ABSTRACT

Mosquitoes are prolific vectors of human pathogens; a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster , is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae ( s.l. ) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti , however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.

4.
bioRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865263

ABSTRACT

The gut epithelium is subject to constant renewal, a process reliant upon intestinal stem cell (ISC) proliferation that is driven by Wnt/ß-catenin signaling. Despite the importance of Wnt signaling within ISCs, the relevance of Wnt signaling within other gut cell types and the underlying mechanisms that modulate Wnt signaling in these contexts remain incompletely understood. Using challenge of the Drosophila midgut with a non-lethal enteric pathogen, we examine the cellular determinants of ISC proliferation, harnessing kramer, a recently identified regulator of Wnt signaling pathways, as a mechanistic tool. We find that Wnt signaling within Prospero-positive cells supports ISC proliferation and that kramer regulates Wnt signaling in this context by antagonizing kelch, a Cullin-3 E3 ligase adaptor that mediates Dishevelled polyubiquitination. This work establishes kramer as a physiological regulator of Wnt/ß-catenin signaling in vivo and suggests enteroendocrine cells as a new cell type that regulates ISC proliferation via Wnt/ß-catenin signaling.

5.
J Virol ; 96(12): e0021522, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35608346

ABSTRACT

The baculovirus envelope protein GP64 is an essential component of the budded virus and is necessary for efficient virion assembly. Little is known regarding intracellular trafficking of GP64 to the plasma membrane, where it is incorporated into budding virions during egress. To identify host proteins and potential cellular trafficking pathways that are involved in delivery of GP64 to the plasma membrane, we developed and characterized a stable Drosophila cell line that inducibly expresses the AcMNPV GP64 protein and used that cell line in combination with a targeted RNA interference (RNAi) screen of vesicular protein trafficking pathway genes. Of the 37 initial hits from the screen, we validated and examined six host genes that were important for trafficking of GP64 to the cell surface. Validated hits included Rab GTPases Rab1 and Rab4, Clathrin heavy chain, clathrin adaptor protein genes AP-1-2ß and AP-2µ, and Snap29. Two gene knockdowns (Rab5 and Exo84) caused substantial increases (up to 2.5-fold) of GP64 on the plasma membrane. We found that a small amount of GP64 is released from cells in exosomes and that some portion of cell surface GP64 is endocytosed, suggesting that recycling helps to maintain GP64 at the cell surface. IMPORTANCE While much is known regarding trafficking of viral envelope proteins in mammalian cells, little is known about this process in insect cells. To begin to understand which factors and pathways are needed for trafficking of insect virus envelope proteins, we engineered a Drosophila melanogaster cell line and implemented an RNAi screen to identify cellular proteins that aid transport of the model baculovirus envelope protein (GP64) to the cell surface. For this we developed an experimental system that leverages the large array of tools available for Drosophila and performed a targeted RNAi screen to identify cellular proteins involved in GP64 trafficking to the cell surface. Since viral envelope proteins are often critical for production of infectious progeny virions, these studies lay the foundation for understanding how either pathogenic insect viruses (baculoviruses) or insect-vectored viruses (e.g., flaviviruses, alphaviruses) egress from cells in tissues such as the midgut to enable systemic virus infection.


Subject(s)
Baculoviridae , Cell Membrane , Insect Proteins , Viral Envelope Proteins , Animals , Baculoviridae/metabolism , Cell Line , Cell Membrane/virology , Drosophila melanogaster/virology , Insect Proteins/genetics , RNA Interference , Viral Envelope Proteins/metabolism
6.
BMC Bioinformatics ; 23(1): 170, 2022 May 09.
Article in English | MEDLINE | ID: mdl-35534830

ABSTRACT

BACKGROUND: Gene co-expression networks (GCNs) can be used to determine gene regulation and attribute gene function to biological processes. Different high throughput technologies, including one and two-channel microarrays and RNA-sequencing, allow evaluating thousands of gene expression data simultaneously, but these methodologies provide results that cannot be directly compared. Thus, it is complex to analyze co-expression relations between genes, especially when there are missing values arising for experimental reasons. Networks are a helpful tool for studying gene co-expression, where nodes represent genes and edges represent co-expression of pairs of genes. RESULTS: In this paper, we establish a method for constructing a gene co-expression network for the Anopheles gambiae transcriptome from 257 unique studies obtained with different methodologies and experimental designs. We introduce the sliding threshold approach to select node pairs with high Pearson correlation coefficients. The resulting network, which we name AgGCN1.0, is robust to random removal of conditions and has similar characteristics to small-world and scale-free networks. Analysis of network sub-graphs revealed that the core is largely comprised of genes that encode components of the mitochondrial respiratory chain and the ribosome, while different communities are enriched for genes involved in distinct biological processes. CONCLUSION: Analysis of the network reveals that both the architecture of the core sub-network and the network communities are based on gene function, supporting the power of the proposed method for GCN construction. Application of network science methodology reveals that the overall network structure is driven to maximize the integration of essential cellular functions, possibly allowing the flexibility to add novel functions.


Subject(s)
Gene Regulatory Networks , Transcriptome , Gene Expression Profiling/methods , Sequence Analysis, RNA
7.
Elife ; 112022 04 26.
Article in English | MEDLINE | ID: mdl-35471187

ABSTRACT

Mosquitoes transmit numerous pathogens, but large gaps remain in our understanding of their physiology. To facilitate explorations of mosquito biology, we have created Aegypti-Atlas (http://aegyptiatlas.buchonlab.com/), an online resource hosting RNAseq profiles of Ae. aegypti body parts (head, thorax, abdomen, gut, Malpighian tubules, ovaries), gut regions (crop, proventriculus, anterior and posterior midgut, hindgut), and a gut time course of blood meal digestion. Using Aegypti-Atlas, we provide insights into regionalization of gut function, blood feeding response, and immune defenses. We find that the anterior and posterior midgut possess digestive specializations which are preserved in the blood-fed state. Blood feeding initiates the sequential induction and repression/depletion of multiple cohorts of peptidases. With respect to defense, immune signaling components, but not recognition or effector molecules, show enrichment in ovaries. Basal expression of antimicrobial peptides is dominated by holotricin and gambicin, which are expressed in carcass and digestive tissues, respectively, in a mutually exclusive manner. In the midgut, gambicin and other effectors are almost exclusively expressed in the anterior regions, while the posterior midgut exhibits hallmarks of immune tolerance. Finally, in a cross-species comparison between Ae. aegypti and Anopheles gambiae midguts, we observe that regional digestive and immune specializations are conserved, indicating that our dataset may be broadly relevant to multiple mosquito species. We demonstrate that the expression of orthologous genes is highly correlated, with the exception of a 'species signature' comprising a few highly/disparately expressed genes. With this work, we show the potential of Aegypti-Atlas to unlock a more complete understanding of mosquito biology.


Subject(s)
Aedes , Anopheles , Aedes/genetics , Animals , Anopheles/genetics , Female , Ovary , Sugars , Transcriptome
8.
Cell Rep ; 38(13): 110572, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35354023

ABSTRACT

Gut microbes play important roles in host physiology; however, the mechanisms underlying their impact remain poorly characterized. Here, we demonstrate that microbes not only influence gut physiology but also alter its epithelial composition. The microbiota and pathogens both influence intestinal stem cell (ISC) differentiation. Intriguingly, while the microbiota promotes ISC differentiation into enterocytes (EC), pathogens stimulate enteroendocrine cell (EE) fate and long-term accumulation of EEs in the midgut epithelium. Importantly, the evolutionarily conserved Drosophila NFKB (Relish) pushes stem cell lineage specification toward ECs by directly regulating differentiation factors. Conversely, the JAK-STAT pathway promotes EE fate in response to infectious damage. We propose a model in which the balance of microbial pattern recognition pathways, such as Imd-Relish, and damage response pathways, such as JAK-STAT, influence ISC differentiation, epithelial composition, and gut physiology.


Subject(s)
Drosophila Proteins , Cell Differentiation/physiology , Drosophila Proteins/metabolism , Enterocytes/metabolism , Intestines , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
9.
Animals (Basel) ; 12(5)2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35268107

ABSTRACT

Marek's disease is an infectious disease in poultry that usually appears in neural and visceral tumors. This disease is caused by Gallid alphaherpesvirus 2 infection in lymphocytes, and its meq gene is commonly used in virulent studies for coding the key protein functional in oncogenic transformation of the lymphocytes. Although vaccines have been introduced in many countries to control its spread and are proven to be efficient, recent records show a decline of such efficiency due to viral evolution. In this study, we reviewed the outbreak of Marek's disease in Asia for the last 10 years, together with associated meq sequences, finding a total of 36 studies recording outbreaks with 132 viral strains in 12 countries. The visceral type is the most common (13 in 16 studies) form of Marek's disease, but additional unobserved neural changes may exist. MD induces liver lymphoma most frequently (11 in 14 studies), and tumors were also found in spleen, kidney, heart, gizzard, skin, intestine, lung, and sciatic nerve. Twelve viral strains distributed in China have been reported to escape the CVI988 vaccine, reaching a mortality rate of more than 30%. Phylogenetic analyses show the internal connection between the Middle East (Turkey, Iraq, Iran, Saudi Arabia), South Asia (India, Indonesia), and East Asia (China and Japan), while external viral communications might occasionally occur. In 18 strains with both sequential and mortality data, amino acid alignment showed several point substitutions that may be related to its virulence. We suggest more behavioral monitoring in Marek's disease-endemic regions and further studies on strain virulence, together with its Meq protein structural changes.

10.
Elife ; 102021 09 23.
Article in English | MEDLINE | ID: mdl-34553686

ABSTRACT

The gut is the primary interface between an animal and food, but how it adapts to qualitative dietary variation is poorly defined. We find that the Drosophila midgut plastically resizes following changes in dietary composition. A panel of nutrients collectively promote gut growth, which sugar opposes. Diet influences absolute and relative levels of enterocyte loss and stem cell proliferation, which together determine cell numbers. Diet also influences enterocyte size. A high sugar diet inhibits translation and uncouples intestinal stem cell proliferation from expression of niche-derived signals, but, surprisingly, rescuing these effects genetically was not sufficient to modify diet's impact on midgut size. However, when stem cell proliferation was deficient, diet's impact on enterocyte size was enhanced, and reducing enterocyte-autonomous TOR signaling was sufficient to attenuate diet-dependent midgut resizing. These data clarify the complex relationships between nutrition, epithelial dynamics, and cell size, and reveal a new mode of plastic, diet-dependent organ resizing.


Subject(s)
Diet , Drosophila melanogaster/growth & development , Gastrointestinal Tract/growth & development , Animals , Animals, Genetically Modified , Cell Proliferation , Drosophila melanogaster/physiology , Enterocytes/cytology , Female , Gastrointestinal Tract/cytology , Gastrointestinal Tract/physiology , Male , Stem Cell Niche
11.
Proc Biol Sci ; 288(1951): 20210786, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34034518

ABSTRACT

A long-standing question in infection biology is why two very similar individuals, with very similar pathogen exposures, may have very different outcomes. Recent experiments have found that even isogenic Drosophila melanogaster hosts, given identical inoculations of some bacterial pathogens at suitable doses, can experience very similar initial bacteria proliferation but then diverge to either a lethal infection or a sustained chronic infection with much lower pathogen load. We hypothesized that divergent infection outcomes are a natural result of mutual negative feedbacks between pathogens and the host immune response. Here, we test this hypothesis in silico by constructing process-based dynamic models for bacterial population growth, host immune induction and the feedbacks between them, based on common mechanisms of immune system response. Mathematical analysis of a minimal conceptual model confirms our qualitative hypothesis that mutual negative feedbacks can magnify small differences among hosts into life-or-death differences in outcome. However, explaining observed features of chronic infections requires an extension of the model to include induced pathogen modifications that shield themselves from host immune responses at the cost of reduced proliferation rate. Our analysis thus generates new, testable predictions about the mechanisms underlying bimodal infection outcomes.


Subject(s)
Drosophila melanogaster , Host-Pathogen Interactions , Animals , Bacteria , Feedback , Immune System
13.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443193

ABSTRACT

Drosophila melanogaster females experience a large shift in energy homeostasis after mating to compensate for nutrient investment in egg production. To cope with this change in metabolism, mated females undergo widespread physiological and behavioral changes, including increased food intake and altered digestive processes. The mechanisms by which the female digestive system responds to mating remain poorly characterized. Here, we demonstrate that the seminal fluid protein Sex Peptide (SP) is a key modulator of female post-mating midgut growth and gene expression. SP is both necessary and sufficient to trigger post-mating midgut growth in females under normal nutrient conditions, and likely acting via its receptor, Sex Peptide Receptor (SPR). Moreover, SP is responsible for almost the totality of midgut transcriptomic changes following mating, including up-regulation of protein and lipid metabolism genes and down-regulation of carbohydrate metabolism genes. These changes in metabolism may help supply the female with the nutrients required to sustain egg production. Thus, we report a role for SP in altering female physiology to enhance reproductive output: Namely, SP triggers the switch from virgin to mated midgut state.


Subject(s)
Digestive System/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fertility/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Peptide/metabolism , Reproduction/physiology , Transcriptome/genetics , Animals , Copulation , Digestive System/anatomy & histology , Digestive System/growth & development , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Fertility/genetics , Gene Ontology , Intestines/anatomy & histology , Intestines/growth & development , Intestines/physiology , Male , Receptors, Peptide/genetics , Reproduction/genetics , Semen/metabolism , Sexual Behavior, Animal/physiology , Transcriptome/physiology
14.
J Innate Immun ; 13(2): 107-126, 2021.
Article in English | MEDLINE | ID: mdl-33207342

ABSTRACT

Insect systemic immune responses to bacterial infections have been mainly studied using microinjections, whereby the microbe is directly injected into the hemocoel. While this methodology has been instrumental in defining immune signaling pathways and enzymatic cascades in the hemolymph, it remains unclear whether and to what extent the contribution of systemic immune defenses to host microbial resistance varies if bacteria invade the hemolymph after crossing the midgut epithelium subsequent to an oral infection. Here, we address this question using the pathogenic Serratia marcescens (Sm) DB11 strain to establish systemic infections of the malaria vector Anopheles gambiae, either by septic Sm injections or by midgut crossing after feeding on Sm. Using functional genetic studies by RNAi, we report that the two humoral immune factors, thioester-containing protein 1 and C-type lectin 4, which play key roles in defense against Gram-negative bacterial infections, are essential for defense against systemic Sm infections established through injection, but they become dispensable when Sm infects the hemolymph following oral infection. Similar results were observed for the mosquito Rel2 pathway. Surprisingly, blocking phagocytosis by cytochalasin D treatment did not affect mosquito susceptibility to Sm infections established through either route. Transcriptomic analysis of mosquito midguts and abdomens by RNA-seq revealed that the transcriptional response in these tissues is more pronounced in response to feeding on Sm. Functional classification of differentially expressed transcripts identified metabolic genes as the most represented class in response to both routes of infection, while immune genes were poorly regulated in both routes. We also report that Sm oral infections are associated with significant downregulation of several immune genes belonging to different families, specifically the clip-domain serine protease family. In sum, our findings reveal that the route of infection not only alters the contribution of key immunity genes to host antimicrobial defense but is also associated with different transcriptional responses in midguts and abdomens, possibly reflecting different adaptive strategies of the host.


Subject(s)
Anopheles/immunology , Hemolymph/immunology , Malaria/immunology , Serratia Infections/immunology , Serratia marcescens/physiology , Animals , Cells, Cultured , Disease Vectors , Down-Regulation , Feeding Methods , Female , Gene Expression Profiling , Immunity, Innate , Insect Proteins/metabolism , Lectins, C-Type/metabolism , Serine Proteases/genetics , Signal Transduction
15.
Pest Manag Sci ; 77(4): 1594-1606, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33342014

ABSTRACT

BACKGROUND: Drosophila suzukii (Matsumura, 1931) (spotted wing drosophila), an invasive species, has recently become a significant global pest of soft-skinned fruits such as berries. Unlike other Drosophila species, female D. suzukii have evolved a specialized sharp, serrated ovipositor that pierces and penetrates ripe and ripening fruits, causing them to lose commercial value and preventing their sale. A first step for the development of biological control agents for pest management may be achieved through the identification of microbes infectious for D. suzukii in the wild. RESULTS: We first determined that D. suzukii is susceptible to chemicals commonly used to rear Drosophilids in the laboratory and established a diet able to sustain healthy D. suzukii growth. Using this diet, we demonstrated that of 25 species of culturable bacteria and fungi isolated from field-collected D. suzukii, eight microbes decreased host survival when injected. Three of the eight bacteria (Alcaligenes faecalis, Achromobacter spanius and Serratia marcescens) were acutely pathogenic to both D. suzukii and Drosophila melanogaster adults by injection. Feeding of these bacteria resulted in susceptibility only in larvae. CONCLUSION: We successfully identified multiple microbes from field-collected D. suzukii that are pathogenic to both larvae and adults through different routes of infection, some of which could be candidates for biocontrol of this species. © 2020 Society of Chemical Industry.


Subject(s)
Achromobacter , Drosophila , Animals , Drosophila melanogaster , Female , Fruit
16.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Article in English | MEDLINE | ID: mdl-31756561

ABSTRACT

BACKGROUND & AIMS: The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis. METHODS: We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice. RESULTS: First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling. CONCLUSIONS: This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.


Subject(s)
Enteroendocrine Cells/physiology , Inhibitor of Apoptosis Proteins/genetics , Intestinal Mucosa/physiology , MicroRNAs/metabolism , Stem Cells/physiology , Animals , Cell Lineage/genetics , Cell Proliferation/genetics , Cells, Cultured , Computational Biology , ErbB Receptors/metabolism , Feeding Behavior/physiology , Female , Inhibitor of Apoptosis Proteins/metabolism , Intestinal Mucosa/cytology , Male , Mice , Mice, Transgenic , Models, Animal , Organoids , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis
17.
Pestic Biochem Physiol ; 161: 95-103, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31685202

ABSTRACT

Insecticides are valuable and widely used tools for the control of pest insects. Despite the use of synthetic insecticides for >50 years, we continue to have a limited understanding of the genes that influence the key steps of the poisoning process. Major barriers for improving our understanding of insecticide toxicity have included a narrow range of tools and/or a large number of candidate genes that could be involved in the poisoning process. Herein, we discuss the numerous tools and resources available in Drosophila melanogaster that could be brought to bear to improve our understanding of the processes determining insecticide toxicity. These include unbiased approaches such as forward genetic screens, population genetic methods and candidate gene approaches. Examples are provided to showcase how D. melanogaster has been successfully used for insecticide toxicology studies in the past, and ideas for future studies using this valuable insect are discussed.


Subject(s)
Drosophila melanogaster/drug effects , Insecticides/toxicity , Animals , Drosophila melanogaster/genetics , Genes, Insect , Insecticides/pharmacokinetics , Models, Genetic
18.
Immunity ; 51(4): 625-637.e3, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31564469

ABSTRACT

Preventing aberrant immune responses against the microbiota is essential for the health of the host. Microbiota-shed pathogen-associated molecular patterns translocate from the gut lumen into systemic circulation. Here, we examined the role of hemolymph (insect blood) filtration in regulating systemic responses to microbiota-derived peptidoglycan. Drosophila deficient for the transcription factor Klf15 (Klf15NN) are viable but lack nephrocytes-cells structurally and functionally homologous to the glomerular podocytes of the kidney. We found that Klf15NN flies were more resistant to infection than wild-type (WT) counterparts but exhibited a shortened lifespan. This was associated with constitutive Toll pathway activation triggered by excess peptidoglycan circulating in Klf15NN flies. In WT flies, peptidoglycan was removed from systemic circulation by nephrocytes through endocytosis and subsequent lysosomal degradation. Thus, renal filtration of microbiota-derived peptidoglycan maintains immune homeostasis in Drosophila, a function likely conserved in mammals and potentially relevant to the chronic immune activation seen in settings of impaired blood filtration.


Subject(s)
Bacterial Infections/immunology , Connective Tissue/physiology , Drosophila/physiology , Kidney Glomerulus/physiology , Kruppel-Like Transcription Factors/genetics , Nuclear Proteins/genetics , Podocytes/physiology , Animals , Animals, Genetically Modified , Bodily Secretions , Drosophila Proteins/metabolism , Endocytosis , Homeostasis , Immunity, Innate , Mammals , Microbiota , Toll-Like Receptors/metabolism
19.
Cell Host Microbe ; 26(3): 412-425.e5, 2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31492656

ABSTRACT

Surviving infection requires immune and repair mechanisms. Developing organisms face the additional challenge of integrating these mechanisms with tightly controlled developmental processes. The larval Drosophila midgut lacks dedicated intestinal stem cells. We show that, upon infection, larvae perform limited repair using adult midgut precursors (AMPs). AMPs differentiate in response to damage to generate new enterocytes, transiently depleting their pool. Developmental delay allows for AMP reconstitution, ensuring the completion of metamorphosis. Notch signaling is required for the differentiation of AMPs into the encasing, niche-like peripheral cells (PCs), but not to differentiate PCs into enterocytes. Dpp (TGF-ß) signaling is sufficient, but not necessary, to induce PC differentiation into enterocytes. Infection-induced JAK-STAT pathway is both required and sufficient for differentiation of AMPs and PCs into new enterocytes. Altogether, this work highlights the constraints imposed by development on an organism's response to infection and demonstrates the transient use of adult precursors for tissue repair.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/metabolism , Gastrointestinal Tract/metabolism , Larva/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Drosophila/microbiology , Drosophila/physiology , Drosophila Proteins/genetics , Enterocytes/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/pathology , Janus Kinases/metabolism , Larva/immunology , Larva/microbiology , Metamorphosis, Biological , Pectobacterium carotovorum/pathogenicity , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcriptome
20.
Wiley Interdiscip Rev Dev Biol ; 8(5): e344, 2019 09.
Article in English | MEDLINE | ID: mdl-30993906

ABSTRACT

From flies to humans, many components of the innate immune system have been conserved during metazoan evolution. This foundational observation has allowed us to develop Drosophila melanogaster, the fruit fly, into a powerful model to study innate immunity in animals. Thanks to an ever-growing arsenal of genetic tools, an easily manipulated genome, and its winning disposition, Drosophila is now employed to study not only basic molecular mechanisms of pathogen recognition and immune signaling, but also the nature of physiological responses activated in the host by microbial challenge and how dysregulation of these processes contributes to disease. Here, we present a collection of methods and protocols to challenge the fly with an assortment of microbes, both systemically and orally, and assess its humoral, cellular, and epithelial response to infection. Our review covers techniques for measuring the reaction to microbial infection both qualitatively and quantitatively. Specifically, we describe survival, bacterial load, BLUD (a measure of disease tolerance), phagocytosis, melanization, clotting, and ROS production assays, as well as efficient protocols to collect hemolymph and measure immune gene expression. We also offer an updated catalog of online resources and a collection of popular reporter lines and mutants to facilitate research efforts. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/complications , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Immunity, Innate/immunology , Phagocytosis , Animals , Bacteria/immunology , Bacterial Infections/immunology , Bacterial Infections/microbiology , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Phenotype , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...