1
|
Deichsel S, Frankenreiter L, Fechner J, Gahr BM, Zimmermann M, Mastel H, Preis I, Preiss A, Nagel AC. Inhibition of the Notch signal transducer CSL by Pkc53E-mediated phosphorylation to fend off parasitic immune challenge in Drosophila. eLife 2024; 12:RP89582. [PMID: 39503739 PMCID: PMC11540305 DOI: 10.7554/elife.89582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Notch signalling activity regulates hematopoiesis in Drosophila and vertebrates alike. Parasitoid wasp infestation of Drosophila larvae, however, requires a timely downregulation of Notch activity to allow the formation of encapsulation-active blood cells. Here, we show that the Drosophila CSL transcription factor Suppressor of Hairless [Su(H)] is phosphorylated at Serine 269 in response to parasitoid wasp infestation. As this phosphorylation interferes with the DNA binding of Su(H), it reversibly precludes its activity. Accordingly, phospho-deficient Su(H)S269A mutants are immune-compromised. A screen for kinases involved in Su(H) phosphorylation identified Pkc53E, required for normal hematopoiesis as well as for parasitoid immune response. Genetic and molecular interactions support the specificity of the Su(H)-Pkc53E relationship. Moreover, phorbol ester treatment inhibits Su(H) activity in vivo and in human cell culture. We conclude that Pkc53E targets Su(H) during parasitic wasp infestation, thereby remodelling the blood cell population required for wasp egg encapsulation.
Collapse
Affiliation(s)
- Sebastian Deichsel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Medical Genetics and Applied Genomics, University of TübingenTübingenGermany
| | - Lisa Frankenreiter
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Johannes Fechner
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Institute of Biomedical Genetics (IBMG), University of StuttgartStuttgartGermany
| | - Bernd M Gahr
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Internal Medicine II, Molecular Cardiology, University of UlmUlmGermany
| | - Mirjam Zimmermann
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Helena Mastel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Irina Preis
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anette Preiss
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anja C Nagel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| |
Collapse
|
2
|
Goins LM, Girard JR, Mondal BC, Buran S, Su CC, Tang R, Biswas T, Kissi JA, Banerjee U. Wnt signaling couples G2 phase control with differentiation during hematopoiesis in Drosophila. Dev Cell 2024; 59:2477-2496.e5. [PMID: 38866012 PMCID: PMC11421984 DOI: 10.1016/j.devcel.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 03/27/2024] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
During homeostasis, a critical balance is maintained between myeloid-like progenitors and their differentiated progeny, which function to mitigate stress and innate immune challenges. The molecular mechanisms that help achieve this balance are not fully understood. Using genetic dissection in Drosophila, we show that a Wnt6/EGFR-signaling network simultaneously controls progenitor growth, proliferation, and differentiation. Unlike G1-quiescence of stem cells, hematopoietic progenitors are blocked in G2 phase by a β-catenin-independent (Wnt/STOP) Wnt6 pathway that restricts Cdc25 nuclear entry and promotes cell growth. Canonical β-catenin-dependent Wnt6 signaling is spatially confined to mature progenitors through localized activation of the tyrosine kinases EGFR and Abelson kinase (Abl), which promote nuclear entry of β-catenin and facilitate exit from G2. This strategy combines transcription-dependent and -independent forms of both Wnt6 and EGFR pathways to create a direct link between cell-cycle control and differentiation. This unique combinatorial strategy employing conserved components may underlie homeostatic balance and stress response in mammalian hematopoiesis.
Collapse
Affiliation(s)
- Lauren M Goins
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Juliet R Girard
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Bama Charan Mondal
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sausan Buran
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chloe C Su
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ruby Tang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Titash Biswas
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jessica A Kissi
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Utpal Banerjee
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Hachfi S, Brun-Barale A, Fichant A, Munro P, Nawrot-Esposito MP, Michel G, Ruimy R, Rousset R, Bonis M, Boyer L, Gallet A. Ingestion of Bacillus cereus spores dampens the immune response to favor bacterial persistence. Nat Commun 2024; 15:7733. [PMID: 39231950 PMCID: PMC11375157 DOI: 10.1038/s41467-024-51689-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024] Open
Abstract
Strains of the Bacillus cereus (Bc) group are sporulating bacteria commonly associated with foodborne outbreaks. Spores are dormant cells highly resistant to extreme conditions. Nevertheless, the pathological processes associated with the ingestion of either vegetative cells or spores remain poorly understood. Here, we demonstrate that while ingestion of vegetative bacteria leads to their rapid elimination from the intestine of Drosophila melanogaster, a single ingestion of spores leads to the persistence of bacteria for at least 10 days. We show that spores do not germinate in the anterior part of the intestine which bears the innate immune defenses. Consequently, spores reach the posterior intestine where they germinate and activate both the Imd and Toll immune pathways. Unexpectedly, this leads to the induction of amidases, which are negative regulators of the immune response, but not to antimicrobial peptides. Thereby, the local germination of spores in the posterior intestine dampens the immune signaling that in turn fosters the persistence of Bc bacteria. This study provides evidence for how Bc spores hijack the intestinal immune defenses allowing the localized birth of vegetative bacteria responsible for the digestive symptoms associated with foodborne illness outbreaks.
Collapse
Affiliation(s)
- Salma Hachfi
- Université Côte d'Azur, CNRS, INRAE, ISA, Sophia Antipolis, France
- Université Côte d'Azur, Inserm, C3M, Nice, France
| | | | - Arnaud Fichant
- Université Côte d'Azur, CNRS, INRAE, ISA, Sophia Antipolis, France
- Anses (Laboratoire de Sécurité des Aliments), Université Paris-Est, Maisons-Alfort, France
| | | | | | | | - Raymond Ruimy
- Université Côte d'Azur, Inserm, C3M, Nice, France
- Bacteriology Laboratory, Archet 2 Hospital, CHU, Université Côte d'Azur, Nice, France
| | - Raphaël Rousset
- Université Côte d'Azur, CNRS, INRAE, ISA, Sophia Antipolis, France
| | - Mathilde Bonis
- Anses (Laboratoire de Sécurité des Aliments), Université Paris-Est, Maisons-Alfort, France
| | | | - Armel Gallet
- Université Côte d'Azur, CNRS, INRAE, ISA, Sophia Antipolis, France.
| |
Collapse
|
4
|
Li Q, Zhang M, Qin S, Wen J, Shen X, Du Z. Dual oxidase 2 (duox 2) participates in the intestinal antibacterial innate immune responses of Procambarus clarkii by regulating ROS levels. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 153:105116. [PMID: 38101716 DOI: 10.1016/j.dci.2023.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/09/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
Dual oxidase (Duox) a member of the nicotinamide adenine dinucleotide phosphate oxidase (NOX) family can induce the production of reactive oxygen species (ROS). In vertebrates, the duox gene was indicated to be associated with the mucosal immunity. The roles of the duox gene in invertebrates were mainly studied in insects for the function of maintaining intestinal flora balance. In recent years, some studies have reported that Duox is involved in regulating the production of ROS and plays an important role in defending against the intestinal pathogen infection. However, the molecular mechanism has not been fully illuminated. In this study, a duox 2 involved in the production of H2O2 was identified for the first time in P. clarkii. Mature Pc-Duox 2 is a 7-transmembrane protein molecule that includes PHD, FAD, and NAD domains. Pc-duox 2 was mainly expressed in hemocytes and intestinal tissue. Its expression levels were obviously upregulated after intramuscular or oral infection with V. harveyi. In the RNAi assay, the upregulated trends of H2O2 and total ROS levels in crayfish intestine were significantly suppressed when Pc-duox 2 was knocked down. Compared with the slightly affected SOD activity, the upregulated CAT activity was suppressed more obviously in the crayfish intestine. Furthermore, Pc-duox 2 had an important effect on the maintenance of the structural stability of crayfish the intestine. Further research revealed that the knockdown of Pc-duox 2 could cause an obvious suppression in the upregulated levels of Toll signalling pathway-related genes, including Pc-toll 1, Pc-toll 3, Pc-dorsal, Pc-ALF 5, Pc-crustin 1, and Pc-lysozyme. Ultimately, these changes triggered the accelerated death of crayfish. Overall, we speculated that Pc-duox 2 played an important role in antibacterial innate immunity in the crayfish intestine by regulating the total ROS level.
Collapse
Affiliation(s)
- Qianqian Li
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China
| | - Mingda Zhang
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China
| | - Shiyu Qin
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China
| | - Jing Wen
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China
| | - Xiuli Shen
- Library, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China
| | - Zhiqiang Du
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia autonomous region, 014010, China.
| |
Collapse
|
5
|
Ji Y, Gao B, Zhao D, Wang Y, Zhang L, Wu H, Xie Y, Shi Q, Guo W. Involvement of Sep38β in the Insecticidal Activity of Bacillus thuringiensis against Beet Armyworm, Spodoptera exigua (Lepidoptera). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2321-2333. [PMID: 38206329 DOI: 10.1021/acs.jafc.3c06667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
The p38 mitogen-activated protein kinases (MAPKs) are associated with insect immunity, tissue repair, and the insecticidal activity of Bacillus thuringiensis (Bt). Here, a p38 MAPK family gene (Sep38β) was identified from Spodoptera exigua. Among the developmental stages, the transcription level of Sep38β was the highest in egg, followed by that in prepupa and pupa. Sep38β expression peaked in Malpighian tubules and the hemolymph of fifth instar larvae. Knockdown of Sep38β or injection of SB203580 (a p38 MAPK inhibitor) significantly downregulated the SeDUOX expression and reactive oxygen species (ROS) level in the midgut, accounting for deterioration of the midgut to scavenge pathogens and enhancement of Bt insecticidal activity. In conclusion, all the results demonstrate that Sep38β regulates the immune-related ROS level in the insect midgut, which suppresses the insecticidal activity of Bt against S. exigua by 17-22%. Our study highlights that Sep38β is essential for insect immunity and the insecticidal activity of Bt to S. exigua and is a potential target for pest control.
Collapse
Affiliation(s)
- Yujie Ji
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Bo Gao
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Dan Zhao
- College of Plant Protection, Hebei Agricultural University, Baoding 071001, China
| | - Yao Wang
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Lu Zhang
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Han Wu
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yifan Xie
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qiuyu Shi
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wei Guo
- Graduate School of Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
6
|
Ma M, Luo J, Li C, Eleftherianos I, Zhang W, Xu L. A life-and-death struggle: interaction of insects with entomopathogenic fungi across various infection stages. Front Immunol 2024; 14:1329843. [PMID: 38259477 PMCID: PMC10800808 DOI: 10.3389/fimmu.2023.1329843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Insects constitute approximately 75% of the world's recognized fauna, with the majority of species considered as pests. Entomopathogenic fungi (EPF) are parasitic microorganisms capable of efficiently infecting insects, rendering them potent biopesticides. In response to infections, insects have evolved diverse defense mechanisms, prompting EPF to develop a variety of strategies to overcome or circumvent host defenses. While the interaction mechanisms between EPF and insects is well established, recent findings underscore that their interplay is more intricate than previously thought, especially evident across different stages of EPF infection. This review primarily focuses on the interplay between EPF and the insect defense strategies, centered around three infection stages: (1) Early infection stage: involving the pre-contact detection and avoidance behavior of EPF in insects, along with the induction of behavioral responses upon contact with the host cuticle; (2) Penetration and intra-hemolymph growth stage: involving the initiation of intricate cellular and humoral immune functions in insects, while symbiotic microbes can further contribute to host resistance; (3) Host insect's death stage: involving the ultimate confrontation between pathogens and insects. Infected insects strive to separate themselves from the healthy population, while pathogens rely on the infected insects to spread to new hosts. Also, we discuss a novel pest management strategy underlying the cooperation between EPF infection and disturbing the insect immune system. By enhancing our understanding of the intricate interplay between EPF and the insect, this review provides novel perspectives for EPF-mediated pest management and developing effective fungal insecticides.
Collapse
Affiliation(s)
- Meiqi Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jing Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Chong Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States
| | - Wei Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering (Ministry of Education), Guizhou University, Guiyang, China
| | - Letian Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
7
|
Zeng T, Fu Q, Luo F, Dai J, Fu R, Qi Y, Deng X, Lu Y, Xu Y. Lactic acid bacteria modulate the CncC pathway to enhance resistance to β-cypermethrin in the oriental fruit fly. THE ISME JOURNAL 2024; 18:wrae058. [PMID: 38618721 PMCID: PMC11069359 DOI: 10.1093/ismejo/wrae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
The gut microbiota of insects has been shown to regulate host detoxification enzymes. However, the potential regulatory mechanisms involved remain unknown. Here, we report that gut bacteria increase insecticide resistance by activating the cap "n" collar isoform-C (CncC) pathway through enzymatically generated reactive oxygen species (ROS) in Bactrocera dorsalis. We demonstrated that Enterococcus casseliflavus and Lactococcus lactis, two lactic acid-producing bacteria, increase the resistance of B. dorsalis to β-cypermethrin by regulating cytochrome P450 (P450) enzymes and α-glutathione S-transferase (GST) activities. These gut symbionts also induced the expression of CncC and muscle aponeurosis fibromatosis. BdCncC knockdown led to a decrease in resistance caused by gut bacteria. Ingestion of the ROS scavenger vitamin C in resistant strain affected the expression of BdCncC/BdKeap1/BdMafK, resulting in reduced P450 and GST activity. Furthermore, feeding with E. casseliflavus or L. lactis showed that BdNOX5 increased ROS production, and BdNOX5 knockdown affected the expression of the BdCncC/BdMafK pathway and detoxification genes. Moreover, lactic acid feeding activated the ROS-associated regulation of P450 and GST activity. Collectively, our findings indicate that symbiotic gut bacteria modulate intestinal detoxification pathways by affecting physiological biochemistry, thus providing new insights into the involvement of insect gut microbes in the development of insecticide resistance.
Collapse
Affiliation(s)
- Tian Zeng
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Qianyan Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Fangyi Luo
- Guangdong Provincial Sericulture & Mulberry Engineering Research Center, Guangdong Prov Key Lab of AgroAnimal Genomics & Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jian Dai
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Rong Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Yixiang Qi
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Xiaojuan Deng
- Guangdong Provincial Sericulture & Mulberry Engineering Research Center, Guangdong Prov Key Lab of AgroAnimal Genomics & Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongyue Lu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| | - Yijuan Xu
- Guangdong Laboratory for Lingnan Modern Agriculture, Department of Entomology, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
8
|
Khan SA, Kojour MAM, Han YS. Recent trends in insect gut immunity. Front Immunol 2023; 14:1272143. [PMID: 38193088 PMCID: PMC10773798 DOI: 10.3389/fimmu.2023.1272143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
The gut is a crucial organ in insect defense against various pathogens and harmful substances in their environment and diet. Distinct insect gut compartments possess unique functionalities contributing to their physiological processes, including immunity. The insect gut's cellular composition is vital for cellular and humoral immunity. The peritrophic membrane, mucus layer, lumen, microvilli, and various gut cells provide essential support for activating and regulating immune defense mechanisms. These components also secrete molecules and enzymes that are imperative in physiological activities. Additionally, the gut microbiota initiates various signaling pathways and produces vitamins and minerals that help maintain gut homeostasis. Distinct immune signaling pathways are activated within the gut when insects ingest pathogens or hazardous materials. The pathway induced depends on the infection or pathogen type; include immune deficiency (imd), Toll, JAK/STAT, Duox-ROS, and JNK/FOXO regulatory pathways. These pathways produce different antimicrobial peptides (AMPs) and maintain gut homeostasis. Furthermore, various signaling mechanisms within gut cells regulate insect gut recovery following infection. Although some questions regarding insect gut immunity in different species require additional study, this review provides insights into the insect gut's structure and composition, commensal microorganism roles in Drosophila melanogaster and Tenebrio molitor life cycles, different signaling pathways involved in gut immune systems, and the insect gut post-infection recovery through various signaling mechanisms.
Collapse
Affiliation(s)
- Shahidul Ahmed Khan
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Maryam Ali Mohmmadie Kojour
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit, University of Bonn, Bonn, Germany
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
9
|
Vertacnik KL, Herrig DK, Godfrey RK, Hill T, Geib SM, Unckless RL, Nelson DR, Linnen CR. Evolution of five environmentally responsive gene families in a pine-feeding sawfly, Neodiprion lecontei (Hymenoptera: Diprionidae). Ecol Evol 2023; 13:e10506. [PMID: 37791292 PMCID: PMC10542623 DOI: 10.1002/ece3.10506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 10/05/2023] Open
Abstract
A central goal in evolutionary biology is to determine the predictability of adaptive genetic changes. Despite many documented cases of convergent evolution at individual loci, little is known about the repeatability of gene family expansions and contractions. To address this void, we examined gene family evolution in the redheaded pine sawfly Neodiprion lecontei, a noneusocial hymenopteran and exemplar of a pine-specialized lineage evolved from angiosperm-feeding ancestors. After assembling and annotating a draft genome, we manually annotated multiple gene families with chemosensory, detoxification, or immunity functions before characterizing their genomic distributions and molecular evolution. We find evidence of recent expansions of bitter gustatory receptor, clan 3 cytochrome P450, olfactory receptor, and antimicrobial peptide subfamilies, with strong evidence of positive selection among paralogs in a clade of gustatory receptors possibly involved in the detection of bitter compounds. In contrast, these gene families had little evidence of recent contraction via pseudogenization. Overall, our results are consistent with the hypothesis that in response to novel selection pressures, gene families that mediate ecological interactions may expand and contract predictably. Testing this hypothesis will require the comparative analysis of high-quality annotation data from phylogenetically and ecologically diverse insect species and functionally diverse gene families. To this end, increasing sampling in under-sampled hymenopteran lineages and environmentally responsive gene families and standardizing manual annotation methods should be prioritized.
Collapse
Affiliation(s)
- Kim L. Vertacnik
- Department of EntomologyUniversity of KentuckyLexingtonKentuckyUSA
| | | | - R. Keating Godfrey
- McGuire Center for Lepidoptera and Biodiversity, University of FloridaGainesvilleFloridaUSA
| | - Tom Hill
- National Institute of Allergy and Infectious DiseasesBethesdaMarylandUSA
| | - Scott M. Geib
- Tropical Crop and Commodity Protection Research UnitUnited States Department of Agriculture: Agriculture Research Service Pacific Basin Agricultural Research CenterHiloHawaiiUSA
| | - Robert L. Unckless
- Department of Molecular BiosciencesUniversity of KansasLawrenceKansasUSA
| | - David R. Nelson
- Department of Microbiology, Immunology and BiochemistryUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | | |
Collapse
|
10
|
Li S, Wang J, Tian X, Toufeeq S, Huang W. Immunometabolic regulation during the presence of microorganisms and parasitoids in insects. Front Immunol 2023; 14:905467. [PMID: 37818375 PMCID: PMC10560992 DOI: 10.3389/fimmu.2023.905467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Multicellular organisms live in environments containing diverse nutrients and a wide variety of microbial communities. On the one hand, the immune response of organisms can protect from the intrusion of exogenous microorganisms. On the other hand, the dynamic coordination of anabolism and catabolism of organisms is a necessary factor for growth and reproduction. Since the production of an immune response is an energy-intensive process, the activation of immune cells is accompanied by metabolic transformations that enable the rapid production of ATP and new biomolecules. In insects, the coordination of immunity and metabolism is the basis for insects to cope with environmental challenges and ensure normal growth, development and reproduction. During the activation of insect immune tissues by pathogenic microorganisms, not only the utilization of organic resources can be enhanced, but also the activated immune cells can usurp the nutrients of non-immune tissues by generating signals. At the same time, insects also have symbiotic bacteria in their body, which can affect insect physiology through immune-metabolic regulation. This paper reviews the research progress of insect immune-metabolism regulation from the perspective of insect tissues, such as fat body, gut and hemocytes. The effects of microorganisms (pathogenic bacteria/non-pathogenic bacteria) and parasitoids on immune-metabolism were elaborated here, which provide guidance to uncover immunometabolism mechanisms in insects and mammals. This work also provides insights to utilize immune-metabolism for the formulation of pest control strategies.
Collapse
Affiliation(s)
- Shirong Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- College of Life Sciences, Yan’an University, Yan’an, Shaanxi, China
| | - Jing Wang
- College of Life Sciences, Shangrao Normal University, Shangrao, China
| | - Xing Tian
- College of Life Sciences, Yan’an University, Yan’an, Shaanxi, China
| | - Shahzad Toufeeq
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Wuren Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
11
|
Qush A, Al Khatib HA, Rachid H, Al-Tamimi H, Al-Eshaq A, Al-Adwi S, Yassine HM, Kamareddine L. Intake of caffeine containing sugar diet remodels gut microbiota and perturbs Drosophila melanogaster immunity and lifespan. Microbes Infect 2023; 25:105149. [PMID: 37169244 DOI: 10.1016/j.micinf.2023.105149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
The diet-microbiome-immunity axis is one among the many arms that draw up the "we are what we intake" proclamation. As such, studies on the effect of food and beverage intake on the gut environment and microbiome and on modulating immunological responses and the host's susceptibility to pathogens are on the rise. A typical accompaniment in different sustenance we consume on daily basis is the trimethylxanthine alkaloid caffeine. Being a chief component in our regular aliment, a better understanding of the effect of caffeine containing food and beverages on our gut-microbiome-immunity axis and henceforth on our health is much needed. In this study, we shed more light on the effect of oral consumption of caffeine supplemented sugar diet on the gut environment, specifically on the gut microbiota, innate immunity and host susceptibility to pathogens using the Drosophila melanogaster model organism. Our findings reveal that the oral intake of a dose-specific caffeine containing sucrose/agarose sugar diet causes a significant alteration within the fly gut milieu demarcated by microbial dysbiosis and an elevation in the production of reactive oxygen species and expression of immune-deficiency (Imd) pathway-dependent antimicrobial peptide genes. The oral intake of caffeine containing sucrose/agarose sugar diet also renders the flies more susceptible to bacterial infection and shortens their lifespan in both infection and non-infection settings. Our findings set forth additional insight into the potentiality of diet to alter the gut milieu and highlight the importance of dietary control on health.
Collapse
Affiliation(s)
- Abeer Qush
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hebah A Al Khatib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Hajar Rachid
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hend Al-Tamimi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Alyaa Al-Eshaq
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Shaima Al-Adwi
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Layla Kamareddine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
12
|
Joshi M, Viallat-Lieutaud A, Royet J. Role of Rab5 early endosomes in regulating Drosophila gut antibacterial response. iScience 2023; 26:107335. [PMID: 37529104 PMCID: PMC10387576 DOI: 10.1016/j.isci.2023.107335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/31/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Interactions between prokaryotes and eukaryotes require a dialogue between MAMPs and PRRs. In Drosophila, bacterial peptidoglycan is detected by PGRP receptors. While the components of the signaling cascades activated upon PGN/PGRP interactions are well characterized, little is known about the subcellular events that translate these early signaling steps into target gene transcription. Using a Drosophila enteric infection model, we show that gut-associated bacteria can induce the formation of intracellular PGRP-LE aggregates which colocalized with the early endosome marker Rab5. Combining microscopic and RNA-seq analysis, we demonstrate that RNAi inactivation of the endocytosis pathway in the Drosophila gut affects the expression of essential regulators of the NF-κB response leading not only to a disruption of the immune response locally in the gut but also at the systemic level. This work sheds new light on the involvement of the endocytosis pathway in the control of the gut response to intestinal bacterial infection.
Collapse
Affiliation(s)
- Manish Joshi
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Annelise Viallat-Lieutaud
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Julien Royet
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
13
|
Wang Z, Yong H, Zhang S, Liu Z, Zhao Y. Colonization Resistance of Symbionts in Their Insect Hosts. INSECTS 2023; 14:594. [PMID: 37504600 PMCID: PMC10380809 DOI: 10.3390/insects14070594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
The symbiotic microbiome is critical in promoting insect resistance against colonization by exogenous microorganisms. The mechanisms by which symbionts contribute to the host's immune capacity is referred to as colonization resistance. Symbionts can protect insects from exogenous pathogens through a variety of mechanisms, including upregulating the expression of host immune-related genes, producing antimicrobial substances, and competitively excluding pathogens. Concordantly, insects have evolved fine-tuned regulatory mechanisms to avoid overactive immune responses against symbionts or specialized cells to harbor symbionts. Alternatively, some symbionts have evolved special adaptations, such as the formation of biofilms to increase their tolerance to host immune responses. Here, we provide a review of the mechanisms about colonization resistance of symbionts in their insect hosts. Adaptations of symbionts and their insect hosts that may maintain such symbiotic relationships, and the significance of such relationships in the coevolution of symbiotic systems are also discussed to provide insights into the in-depth study of the contribution of symbionts to host physiology and behavior.
Collapse
Affiliation(s)
- Zhengyan Wang
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Hanzi Yong
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Shan Zhang
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Zhiyuan Liu
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Yaru Zhao
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| |
Collapse
|
14
|
Loudhaief R, Jneid R, Christensen CF, Mackay DJ, Andersen DS, Colombani J. The Drosophila tumor necrosis factor receptor, Wengen, couples energy expenditure with gut immunity. SCIENCE ADVANCES 2023; 9:eadd4977. [PMID: 37294765 DOI: 10.1126/sciadv.add4977] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 05/04/2023] [Indexed: 06/11/2023]
Abstract
It is well established that tumor necrosis factor (TNF) plays an instrumental role in orchestrating the metabolic disorders associated with late stages of cancers. However, it is not clear whether TNF/TNF receptor (TNFR) signaling controls energy homeostasis in healthy individuals. Here, we show that the highly conserved Drosophila TNFR, Wengen (Wgn), is required in the enterocytes (ECs) of the adult gut to restrict lipid catabolism, suppress immune activity, and maintain tissue homeostasis. Wgn limits autophagy-dependent lipolysis by restricting cytoplasmic levels of the TNFR effector, TNFR-associated factor 3 (dTRAF3), while it suppresses immune processes through inhibition of the dTAK1/TAK1-Relish/NF-κB pathway in a dTRAF2-dependent manner. Knocking down dTRAF3 or overexpressing dTRAF2 is sufficient to suppress infection-induced lipid depletion and immune activation, respectively, showing that Wgn/TNFR functions as an intersection between metabolism and immunity allowing pathogen-induced metabolic reprogramming to fuel the energetically costly task of combatting an infection.
Collapse
Affiliation(s)
- Rihab Loudhaief
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| | - Rouba Jneid
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| | - Christian Fokdal Christensen
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| | - Duncan J Mackay
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| | - Ditte S Andersen
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| | - Julien Colombani
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, room 439, 2100 Copenhagen O, Denmark
| |
Collapse
|
15
|
Zhao D, Wu H, Li Y, Wang Q, Ji Y, Guo X, Guo W. Effects of the pyrE deletion mutant from Bacillus thuringiensis on gut microbiota and immune response of Spodoptera exigua. Front Microbiol 2023; 14:1182699. [PMID: 37333629 PMCID: PMC10272597 DOI: 10.3389/fmicb.2023.1182699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
The gut microbiota is essential for the growth and development of insects, and the intestinal immune system plays a critical role in regulating the homeostasis of intestinal microorganisms and their interactions with pathogenic bacteria. Infection with Bacillus thuringiensis (Bt) can disrupt the gut microbiota of insects, but the regulatory factors governing the interaction between Bt and gut bacteria are not well understood. Uracil secreted by exogenous pathogenic bacteria can activate DUOX-mediated reactive oxygen species (ROS) production, which helps maintain intestinal microbial homeostasis and immune balance. To elucidate the regulatory genes involved in the interaction between Bt and gut microbiota, we investigate the effects of uracil derived from Bt on gut microbiota, and host immunity using a uracil deficient Bt strain (Bt GS57△pyrE) obtained by homologous recombination. We analyze the biological characteristics of the uracil deficient strain and found that the deletion of uracil in Bt GS57 strain changed the diversity of gut bacteria in Spodoptera exigua, as investigated using Illumina HiSeq sequencing. Furthermore, qRT-PCR analysis showed that compared with Bt GS57 (control), the expression of the SeDuox gene and the level of ROS were significantly decreased after feeding with Bt GS57△pyrE. Adding uracil to Bt GS57△pyrE restored the expression level of DUOX and ROS to a higher level. Additionally, we observed that PGRP-SA, attacin, defensin and ceropin genes were significant different in the midgut of S. exigua infected by Bt GS57 and Bt GS57△pyrE, with a trend of increasing first and then decreasing. These results suggest that uracil regulates and activates the DUOX-ROS system, affects the expression of antimicrobial peptide genes, and disturb intestinal microbial homeostasis. We preliminarily speculate that uracil is a key factor in the interaction between Bt and gut microbiota, and these findings provide a theoretical basis for clarifying the interaction between Bt, host, and intestinal microorganisms, as well as for gaining new insights into the insecticidal mechanism of B. thuringiensis in insects.
Collapse
Affiliation(s)
- Dan Zhao
- College of Plant Protection, Hebei Agricultural University, Baoding, China
| | - Han Wu
- College of Plant Protection, Hebei Agricultural University, Baoding, China
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yazi Li
- College of Plant Protection, Hebei Agricultural University, Baoding, China
| | - Qian Wang
- College of Plant Protection, Hebei Agricultural University, Baoding, China
| | - Yujie Ji
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaochang Guo
- College of Plant Protection, Hebei Agricultural University, Baoding, China
| | - Wei Guo
- College of Plant Protection, Hebei Agricultural University, Baoding, China
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
16
|
Bossen J, Kühle JP, Roeder T. The tracheal immune system of insects - A blueprint for understanding epithelial immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103960. [PMID: 37235953 DOI: 10.1016/j.ibmb.2023.103960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
The unique design of respiratory organs in multicellular organisms makes them prone to infection by pathogens. To cope with this vulnerability, highly effective local immune systems evolved that are also operative in the tracheal system of insects. Many pathogens and parasites (including viruses, bacteria, fungi, and metazoan parasites) colonize the trachea or invade the host via this route. Currently, only two modules of the tracheal immune system have been characterized in depth: 1) Immune deficiency pathway-mediated activation of antimicrobial peptide gene expression and 2) local melanization processes that protect the structure from wounding. There is an urgent need to increase our understanding of the architecture of tracheal immune systems, especially regarding those mechanisms that enable the maintenance of immune homeostasis. This need for new studies is particularly exigent for species other than Drosophila.
Collapse
Affiliation(s)
- Judith Bossen
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Jan-Philip Kühle
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany
| | - Thomas Roeder
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
17
|
Zhang X, Shao W, Huo Y, Kong Y, Zhang W, Li S, Zhou W, Wu X, Qin F, Hu X. The effects of short-term dietary exposure to SiO 2 nanoparticle on the domesticated lepidopteran insect model silkworm (Bombyx mori): Evidence from the perspective of multi-omics. CHEMOSPHERE 2023; 323:138257. [PMID: 36868417 DOI: 10.1016/j.chemosphere.2023.138257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/25/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Silicon dioxide nanoparticles (nSiO2) are one of the widely utilized nanoparticle (NPSs) materials, and exposure to nSiO2 is ubiquitous. With the increasing commercialization of nSiO2, the potential risk of nSiO2 release to the health and the ecological environment have been attracted more attention. In this study, the domesticated lepidopteran insect model silkworm (Bombyx mori) was utilized to evaluate the biological effects of dietary exposure to nSiO2. Histological investigations showed that nSiO2 exposure resulted in midgut tissue injury in a dose-dependent manner. Larval body mass and cocoon production were reduced by nSiO2 exposure. ROS burst was not triggered, and the activities of antioxidant enzymes were induced in the midgut of silkworm exposure to nSiO2. RNA-sequencing revealed that the differentially expressed genes induced by nSiO2 exposure were predominantly enriched into xenobiotics biodegradation and metabolism, lipid, and amino acid metabolism pathways. 16 S rDNA sequencing revealed that nSiO2 exposure altered the microbial diversity in the gut of the silkworm. Metabolomics analysis showed that the combined uni- and multivariate analysis identified 28 significant differential metabolites from the OPLS-DA model. These significant differential metabolites were predominantly enriched into the metabolic pathways, including purine metabolism and tyrosine metabolism and so. Spearman correlation analysis and the Sankey diagram established the relationship between microbe and metabolites, and some genera may play crucial and pleiotropic functions in the interaction between microbiome and host. These findings indicated that nSiO2 exposure could impact the dysregulation of genes related to xenobiotics metabolism, gut dysbiosis, and metabolic pathways and provided a valuable reference for assessing nSiO2 toxicity from a multi-dimensional perspective.
Collapse
Affiliation(s)
- Xing Zhang
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Wenjing Shao
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Yiming Huo
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Yifei Kong
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Wenxue Zhang
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Song Li
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China
| | - Wenlin Zhou
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Xuehui Wu
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Fenju Qin
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, 215009, China.
| | - Xiaolong Hu
- School of Biology & Basic Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
18
|
Hrithik MTH, Ahmed S, Kim Y. Damage signal induced by Bacillus thuringiensis infection triggers immune responses via a DAMP molecule in lepidopteran insect, Spodoptera exigua. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104559. [PMID: 36181778 DOI: 10.1016/j.dci.2022.104559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Insect immunity defends the infection of an insect pathogenic bacterium, Bacillus thuringiensis (Bt). However, it was not clear on the recognition of Bt infection by the insect immune system. This study tested a physiological function of dorsal switch protein 1 (DSP1) in the Bt infection. DSP1 is classified into HMGB1-like damage-associated molecular pattern (DAMP) in insects. Upon Bt infection in a lepidopteran Spodoptera exigua, DSP1 was released from the nuclei of the midgut epithelium and activated immune responses. For this DSP1 release, a functional binding between Bt and its receptors on the midgut epithelium was required because any RNA interference (RNAi) treatments of Bt receptor (cadherin or ABCC) prevented the DSP1 release and became susceptible to the bacterial infection. The DSP1 release was required for the gene induction of Repat33, which is a member of response to pathogen gene family and its gene product mediated cellular and humoral immune responses against pathogen infection in S. exigua. The released DSP1 activated phospholipase A2 (PLA2) to produce eicosanoids, which induced the Repat33 expression because a hemocoelic injection of a recombinant DSP1 induced the Repat33 expression without Bt infection. However, any inhibition of PLA2 activity impaired the DAMP signaling between DSP1 and Repat33. DSP1 also up-regulated two other immune mediators, nitric oxide (NO) and a cytokine called plasmatocyte-spreading peptide (PSP). Either NO or PSP activated PLA2 to up-regulate Repat33 expression. These results suggest that Bt infection of the insect midgut generates a DAMP signal via DSP1 release, which turns on NO or the cytokine-PLA2-Repat33 immune signaling pathway.
Collapse
Affiliation(s)
| | - Shabbir Ahmed
- Department of Plant Medicals, Andong National University, Andong, 36729, South Korea
| | - Yonggyun Kim
- Department of Plant Medicals, Andong National University, Andong, 36729, South Korea.
| |
Collapse
|
19
|
Zeng T, Jaffar S, Xu Y, Qi Y. The Intestinal Immune Defense System in Insects. Int J Mol Sci 2022; 23:ijms232315132. [PMID: 36499457 PMCID: PMC9740067 DOI: 10.3390/ijms232315132] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
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.
Collapse
|
20
|
Roy MC, Ahmed S, Kim Y. Dorsal switch protein 1 as a damage signal in insect gut immunity to activate dual oxidase via an eicosanoid, PGE 2. Front Immunol 2022; 13:994626. [PMID: 36439105 PMCID: PMC9691268 DOI: 10.3389/fimmu.2022.994626] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/18/2022] [Indexed: 08/05/2023] Open
Abstract
Various microbiota including beneficial symbionts reside in the insect gut. Infections of pathogens cause dysregulation of the microflora and threaten insect survival. Reactive oxygen species (ROS) have been used in the gut immune responses, in which its production is tightly regulated by controlling dual oxidase (Duox) activity via Ca2+ signal to protect beneficial microflora and gut epithelium due to its high cytotoxicity. However, it was not clear how the insects discriminate the pathogens from the various microbes in the gut lumen to trigger ROS production. An entomopathogenic nematode (Steinernema feltiae) infection elevated ROS level in the gut lumen of a lepidopteran insect, Spodoptera exigua. Dorsal switch protein 1 (DSP1) localized in the nucleus in the midgut epithelium was released into plasma upon the nematode infection and activated phospholipase A2 (PLA2). The activated PLA2 led to an increase of PGE2 level in the midgut epithelium, in which rising Ca2+ signal up-regulated ROS production. Inhibiting DSP1 release by its specific RNA interference (RNAi) or specific inhibitor, 3-ethoxy-4-methoxyphenol, treatment failed to increase the intracellular Ca2+ level and subsequently prevented ROS production upon the nematode infection. A specific PLA2 inhibitor treatment also prevented the up-regulation of Ca2+ and subsequent ROS production upon the nematode infection. However, the addition of PGE2 to the inhibitor treatment rescued the gut immunity. DSP1 release was not observed at infection with non-pathogenic pathogens but detected in plasma with pathogenic infections that would lead to damage to the gut epithelium. These results indicate that DSP1 acts as a damage-associated molecular pattern in gut immunity through DSP1/PLA2/Ca2+/Duox.
Collapse
|
21
|
Zeng LH, Barkat MQ, Syed SK, Shah S, Abbas G, Xu C, Mahdy A, Hussain N, Hussain L, Majeed A, Khan KUR, Wu X, Hussain M. Hedgehog Signaling: Linking Embryonic Lung Development and Asthmatic Airway Remodeling. Cells 2022; 11:1774. [PMID: 35681469 PMCID: PMC9179967 DOI: 10.3390/cells11111774] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 12/28/2022] Open
Abstract
The development of the embryonic lung demands complex endodermal-mesodermal interactions, which are regulated by a variety of signaling proteins. Hedgehog (Hh) signaling is vital for lung development. It plays a key regulatory role during several morphogenic mechanisms, such as cell growth, differentiation, migration, and persistence of cells. On the other hand, abnormal expression or loss of regulation of Hh signaling leads to airway asthmatic remodeling, which is characterized by cellular matrix modification in the respiratory system, goblet cell hyperplasia, deposition of collagen, epithelial cell apoptosis, proliferation, and activation of fibroblasts. Hh also targets some of the pathogens and seems to have a significant function in tissue repairment and immune-related disorders. Similarly, aberrant Hh signaling expression is critically associated with the etiology of a variety of other airway lung diseases, mainly, bronchial or tissue fibrosis, lung cancer, and pulmonary arterial hypertension, suggesting that controlled regulation of Hh signaling is crucial to retain healthy lung functioning. Moreover, shreds of evidence imply that the Hh signaling pathway links to lung organogenesis and asthmatic airway remodeling. Here, we compiled all up-to-date investigations linked with the role of Hh signaling in the development of lungs as well as the attribution of Hh signaling in impairment of lung expansion, airway remodeling, and immune response. In addition, we included all current investigational and therapeutic approaches to treat airway asthmatic remodeling and immune system pathway diseases.
Collapse
Affiliation(s)
- Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China;
| | - Muhammad Qasim Barkat
- Key Laboratory of CFDA for Respiratory Drug Research, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.Q.B.); (C.X.)
| | - Shahzada Khurram Syed
- Department of Basic Medical Sciences, School of Health Sciences, University of Management and Technology Lahore, Lahore 54000, Pakistan;
| | - Shahid Shah
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan; (S.S.); (G.A.); (L.H.)
| | - Ghulam Abbas
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan; (S.S.); (G.A.); (L.H.)
| | - Chengyun Xu
- Key Laboratory of CFDA for Respiratory Drug Research, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.Q.B.); (C.X.)
| | - Amina Mahdy
- Medical Pharmacology Department, International School of Medicine, Istanbul Medipol University, Istanbul 34000, Turkey;
| | - Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain 64141, United Arab Emirates;
| | - Liaqat Hussain
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan; (S.S.); (G.A.); (L.H.)
| | - Abdul Majeed
- Faculty of Pharmacy, Bahauddin Zakariya University, Mulatn 60000, Pakistan;
| | - Kashif-ur-Rehman Khan
- Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China;
| | - Musaddique Hussain
- Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| |
Collapse
|
22
|
Deshpande R, Lee B, Qiao Y, Grewal SS. TOR signalling is required for host lipid metabolic remodelling and survival following enteric infection in Drosophila. Dis Model Mech 2022; 15:dmm049551. [PMID: 35363274 PMCID: PMC9118046 DOI: 10.1242/dmm.049551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 12/29/2022] Open
Abstract
When infected by enteric pathogenic bacteria, animals need to initiate local and whole-body defence strategies. Although most attention has focused on the role of innate immune anti-bacterial responses, less is known about how changes in host metabolism contribute to host defence. Using Drosophila as a model system, we identify induction of intestinal target-of-rapamycin (TOR) kinase signalling as a key adaptive metabolic response to enteric infection. We find that enteric infection induces both local and systemic induction of TOR independently of the Immune deficiency (IMD) innate immune pathway, and we see that TOR functions together with IMD signalling to promote infection survival. These protective effects of TOR signalling are associated with remodelling of host lipid metabolism. Thus, we see that TOR is required to limit excessive infection-mediated wasting of host lipid stores by promoting an increase in the levels of gut- and fat body-expressed lipid synthesis genes. Our data support a model in which induction of TOR represents a host tolerance response to counteract infection-mediated lipid wasting in order to promote survival. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | - Savraj S. Grewal
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
23
|
Nath A, Chakrabarti P, Sen S, Barui A. Reactive Oxygen Species in Modulating Intestinal Stem Cell Dynamics and Function. Stem Cell Rev Rep 2022; 18:2328-2350. [DOI: 10.1007/s12015-022-10377-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 10/18/2022]
|
24
|
Abstract
In adult insects, as in vertebrates, the gut epithelium is a highly regenerative tissue that can renew itself rapidly in response to changing inputs from nutrition, the gut microbiota, ingested toxins, and signals from other organs. Because of its cellular and genetic similarities to the mammalian intestine, and its relevance as a target for the control of insect pests and disease vectors, many researchers have used insect intestines to address fundamental questions about stem cell functions during tissue maintenance and regeneration. In Drosophila, where most of the experimental work has been performed, not only are intestinal cell types and behaviors well characterized, but numerous cell signaling interactions have been detailed that mediate gut epithelial regeneration. A prevailing model for regenerative responses in the insect gut invokes stress sensing by damaged enterocytes (ECs) as a principal source for signaling that activates the division of intestinal stem cells (ISCs) and the growth and differentiation of their progeny. However, extant data also reveal alternative mechanisms for regeneration that involve ISC-intrinsic functions, active culling of healthy epithelial cells, enhanced EC growth, and even cytoplasmic shedding by infected ECs. This article reviews current knowledge of the molecular mechanisms involved in gut regeneration in several insect models (Drosophila and Aedes of the order Diptera, and several Lepidoptera).
Collapse
Affiliation(s)
- Peng Zhang
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| | - Bruce A Edgar
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
25
|
Sensing microbial infections in the Drosophila melanogaster genetic model organism. Immunogenetics 2022; 74:35-62. [DOI: 10.1007/s00251-021-01239-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022]
|
26
|
Telleria EL, Azevedo-Brito DA, Kykalová B, Tinoco-Nunes B, Pitaluga AN, Volf P, Traub-Csekö YM. Leishmania infantum Infection Modulates the Jak-STAT Pathway in Lutzomyia longipalpis LL5 Embryonic Cells and Adult Females, and Affects Parasite Growth in the Sand Fly. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.747820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Phlebotomine sand flies (Diptera, Psychodidae) belonging to the Lutzomyia genus transmit zoonoses in the New World. Lutzomyia longipalpis is the main vector of Leishmania infantum, which is the causative agent of visceral leishmaniasis in Brazil. To identify key molecular aspects involved in the interaction between vector and pathogens and contribute to developing disease transmission controls, we investigated the sand fly innate immunity mediated by the Janus kinase/signal transducer and activator of transcription (Jak-STAT) pathway in response to L. infantum infection. We used two study models: L. longipalpis LL5 embryonic cells co-cultured with L. infantum and sand fly females artificially infected with the parasite. We used qPCR to follow the L. longipalpis gene expression of molecules involved in the Jak-STAT pathway. Also, we modulated the Jak-STAT mediated immune response to understand its role in Leishmania parasite infection. For that, we used RNAi to silence the pathway regulators, protein inhibitor of activated STATs (PIAS) in LL5 cells, and STAT in adult females. In addition, the pathway suppression effect on parasite development within the vector was assessed by light microscopy in late-phase infection. The silencing of the repressor PIAS in LL5 cells led to a moderate increase in a protein tyrosine phosphatase 61F (PTP61F) expression. It suggests a compensatory regulation between these two repressors. L. infantum co-culture with LL5 cells upregulated repressors PIAS, suppressor of cytokine signaling (SOCS), and PTP61F. It also downmodulated virus-induced RNA-1 (VIR-1), a pathway effector, indicating that the parasite could repress the Jak-STAT pathway in LL5 cells. In Leishmania-infected L. longipalpis females, STAT and the antimicrobial peptide attacin were downregulated on the third day post-infection, suggesting a correlation that favors the parasite survival at the end of blood digestion in the sand fly. The antibiotic treatment of infected females showed that the reduction of gut bacteria had little effect on the Jak-STAT pathway regulation. STAT gene silencing mediated by RNAi reduced the expression of inducible nitric oxide synthase (iNOS) and favored Leishmania growth in sand flies on the first day post-infection. These results indicate that STAT participated in the iNOS regulation with subsequent effect on parasite survival.
Collapse
|
27
|
Jahnes BC, Poudel K, Staats AM, Sabree ZL. Microbial colonization promotes model cockroach gut tissue growth and development. JOURNAL OF INSECT PHYSIOLOGY 2021; 133:104274. [PMID: 34216600 DOI: 10.1016/j.jinsphys.2021.104274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Digestive tissues are essential for diet processing and nutrient accessibility, especially in omnivores, and these functions occur despite and in collaboration with dynamic microbial communities that reside within and upon these tissues. Prolonged host development and reduced digestive tissue sizes have been observed in germ-free animals, and normal host phenotypes were recovered following the re-introduction of typical gut microbiomes via coprophagy. RESULTS High-resolution histological analyses of Periplaneta americana cockroach digestive tissues revealed that total prevention of microbial colonization of the gut had severe impacts on the growth and development of gut tissues, especially the posterior midgut and anterior hindgut subcompartments that are expected to be colonized and inhabited by the greatest number of bacteria. Juveniles that were briefly exposed to normal gut microbiota exhibited a partial gut morphological recovery, suggesting that a single inoculation was insufficient. These data highlight gut microbiota as integral to normal growth and development of tissues they are in direct contact with and, more broadly, the organism in which they reside. CONCLUSIONS We draw on these data, host life history traits (i.e. multigenerational cohousing, molting, and filial coprophagy and exuvia feeding), and previous studies to suggest a host developmental model in which gut tissues reflect a conflict-collaboration dynamic where 1) nutrient-absorptive anterior midgut tissues are in competition with transient and resident bacteria for easily assimilable dietary nutrients and whose growth is least-affected by the presence of gut bacteria and 2) posterior midgut, anterior hindgut, and to a lesser degree, posterior hindgut tissues are significantly impacted by gut bacterial presence because they are occupied by the greatest number of bacteria and the host is relying upon, and thus collaborating with, them to assist with complex polysaccharide catabolism processing and nutrient provisioning (i.e. short-chain fatty acids).
Collapse
Affiliation(s)
- Benjamin C Jahnes
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | - Keyshap Poudel
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH, USA
| | - Amelia M Staats
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | - Zakee L Sabree
- Department of Microbiology, Ohio State University, Columbus, OH, USA; Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
28
|
Reactive Oxygen Species-Dependent Innate Immune Mechanisms Control Methicillin-Resistant Staphylococcus aureus Virulence in the Drosophila Larval Model. mBio 2021; 12:e0027621. [PMID: 34126772 PMCID: PMC8262968 DOI: 10.1128/mbio.00276-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Antibiotic-resistant Staphylococcus aureus strains constitute a major public health concern worldwide and are responsible for both health care- and community-associated infections. Here, we establish a robust and easy-to-implement model of oral S. aureus infection using Drosophila melanogaster larvae that allowed us to follow the fate of S. aureus at the whole-organism level as well as the host immune responses. Our study demonstrates that S. aureus infection triggers H2O2 production by the host via the Duox enzyme, thereby promoting antimicrobial peptide production through activation of the Toll pathway. Staphylococcal catalase mediates H2O2 neutralization, which not only promotes S. aureus survival but also minimizes the host antimicrobial response, hence reducing bacterial clearance in vivo. We show that while catalase expression is regulated in vitro by the accessory gene regulatory system (Agr) and the general stress response regulator sigma B (SigB), it no longer depends on these two master regulators in vivo. Finally, we confirm the versatility of this model by demonstrating the colonization and host stimulation capabilities of S. aureus strains belonging to different sequence types (CC8 and CC5) as well as of two other bacterial pathogens, Salmonella enterica serovar Typhimurium and Shigella flexneri. Thus, the Drosophila larva can be a general model to follow in vivo the innate host immune responses triggered during infection by human pathogens.
Collapse
|
29
|
Rahman SR, Terranova T, Tian L, Hines HM. Developmental Transcriptomics Reveals a Gene Network Driving Mimetic Color Variation in a Bumble Bee. Genome Biol Evol 2021; 13:6244266. [PMID: 33881508 PMCID: PMC8220310 DOI: 10.1093/gbe/evab080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2021] [Indexed: 11/24/2022] Open
Abstract
A major goal of evolutionary genetics and evo-devo is to understand how changes in genotype manifest as changes in phenotype. Bumble bees display remarkable color pattern diversity while converging onto numerous regional Müllerian mimicry patterns, thus enabling exploration of the genetic mechanisms underlying convergent phenotypic evolution. In western North America, multiple bumble bee species converge onto local mimicry patterns through parallel shifts of midabdominal segments from red to black. It was previously demonstrated that a Hox gene, Abd-B, is the key regulator of the phenotypic switch in one of these species, Bombus melanopygus, however, the mechanism by which Abd-B regulates color differentiation remains unclear. Using tissue/stage-specific transcriptomic analysis followed by qRT–PCR validation, this study reveals a suite of genes potentially involved downstream of Abd-B during color pattern differentiation. The data support differential genes expression of not only the first switch gene Abd-B, but also an intermediate developmental gene nubbin, and a whole suite of downstream melanin and redox genes that together reinforce the observed eumelanin (black)-pheomelanin (red) ratios. These include potential genes involved in the production of insect pheomelanins, a pigment until recently not thought to occur in insects and thus lacking known regulatory enzymes. The results enhance understanding of pigmentation gene networks involved in bumble bee color pattern development and diversification, while providing insights into how upstream regulators such as Hox genes interact with downstream morphogenic players to facilitate this adaptive phenotypic radiation.
Collapse
Affiliation(s)
- Sarthok Rasique Rahman
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA.,Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Tatiana Terranova
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Li Tian
- Department of Entomology, China Agricultural University, Beijing, China
| | - Heather M Hines
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA.,Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
30
|
Regulators and signalling in insect antimicrobial innate immunity: Functional molecules and cellular pathways. Cell Signal 2021; 83:110003. [PMID: 33836260 DOI: 10.1016/j.cellsig.2021.110003] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 12/29/2022]
Abstract
Insects possess an immune system that protects them from attacks by various pathogenic microorganisms that would otherwise threaten their survival. Immune mechanisms may deal directly with the pathogens by eliminating them from the host organism or disarm them by suppressing the synthesis of toxins and virulence factors that promote the invasion and destructive action of the intruder within the host. Insects have been established as outstanding models for studying immune system regulation because innate immunity can be explored as an integrated system at the level of the whole organism. Innate immunity in insects consists of basal immunity that controls the constitutive synthesis of effector molecules such as antimicrobial peptides, and inducible immunity that is activated after detection of a microbe or its product(s). Activation and coordination of innate immune defenses in insects involve evolutionary conserved immune factors. Previous research in insects has led to the identification and characterization of distinct immune signalling pathways that modulate the response to microbial infections. This work has not only advanced the field of insect immunology, but it has also rekindled interest in the innate immune system of mammals. Here we review the current knowledge on key molecular components of insect immunity and discuss the opportunities they present for confronting infectious diseases in humans.
Collapse
|
31
|
Bai S, Yao Z, Raza MF, Cai Z, Zhang H. Regulatory mechanisms of microbial homeostasis in insect gut. INSECT SCIENCE 2021; 28:286-301. [PMID: 32888254 DOI: 10.1111/1744-7917.12868] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Insects live in incredibly complex environments. The intestinal epithelium of insects is in constant contact with microorganisms, some of which are beneficial and some harmful to the host. Insect gut health and function are maintained through multidimensional mechanisms that can proficiently remove foreign pathogenic microorganisms while effectively maintaining local symbiotic microbial homeostasis. The basic immune mechanisms of the insect gut, such as the dual oxidase-reactive oxygen species (Duox-ROS) system and the immune deficiency (Imd)-signaling pathway, are involved in the maintenance of microbial homeostasis. This paper reviews the role of physical defenses, the Duox-ROS and Imd signaling pathways, the Janus kinase/signal transducers and activators of transcription signaling pathway, and intestinal symbiotic flora in the homeostatic maintenance of the insect gut microbiome.
Collapse
Affiliation(s)
- Shuai Bai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhichao Yao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Fahim Raza
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhaohui Cai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
32
|
Morris O, Jasper H. Reactive Oxygen Species in intestinal stem cell metabolism, fate and function. Free Radic Biol Med 2021; 166:140-146. [PMID: 33600942 DOI: 10.1016/j.freeradbiomed.2021.02.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
Long dismissed as merely harmful respiratory by-products, Reactive Oxygen Species (ROS) have emerged as critical intracellular messengers during cell growth and differentiation. ROS's signaling roles are particularly prominent within the intestine, whose high regenerative capacity is maintained by Intestinal Stem Cells (ISCs). In this review, we outline roles for ROS in ISCs as revealed by studies using Drosophila and mouse model systems. We focus particularly on recent studies highlighting how ROS ties to metabolic adaptations, which ensure energy supply matches demand during ISC activation and differentiation. We describe how declines in these adaptive mechanisms, through aging or pathology, promote reciprocal changes in ISC metabolism and ROS signaling. These changes ultimately contribute to aberrant ISC function, a loss of tissue homeostasis, and a shortened lifespan.
Collapse
Affiliation(s)
- Otto Morris
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945-1400, USA.
| |
Collapse
|
33
|
Madi JR, Outa AA, Ghannam M, Hussein HM, Shehab M, Hasan ZAKH, Fayad AA, Shirinian M, Rahal EA. Drosophila melanogaster as a Model System to Assess the Effect of Epstein-Barr Virus DNA on Inflammatory Gut Diseases. Front Immunol 2021; 12:586930. [PMID: 33828545 PMCID: PMC8019809 DOI: 10.3389/fimmu.2021.586930] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 03/03/2021] [Indexed: 01/05/2023] Open
Abstract
The Epstein-Barr virus (EBV) commonly infects humans and is highly associated with different types of cancers and autoimmune diseases. EBV has also been detected in inflamed gastrointestinal mucosa of patients suffering from prolonged inflammation of the digestive tract such as inflammatory bowel disease (IBD) with no clear role identified yet for EBV in the pathology of such diseases. Since we have previously reported immune-stimulating capabilities of EBV DNA in various models, in this study we investigated whether EBV DNA may play a role in exacerbating intestinal inflammation through innate immune and regeneration responses using the Drosophila melanogaster model. We have generated inflamed gastrointestinal tracts in adult fruit flies through the administration of dextran sodium sulfate (DSS), a sulfated polysaccharide that causes human ulcerative colitis- like pathologies due to its toxicity to intestinal cells. Intestinal damage induced by inflammation recruited plasmatocytes to the ileum in fly hindguts. EBV DNA aggravated inflammation by enhancing the immune deficiency (IMD) pathway as well as further increasing the cellular inflammatory responses manifested upon the administration of DSS. The study at hand proposes a possible immunostimulatory role of the viral DNA exerted specifically in the fly hindgut hence further developing our understanding of immune responses mounted against EBV DNA in the latter intestinal segment of the D. melanogaster gut. These findings suggest that EBV DNA may perpetuate proinflammatory processes initiated in an inflamed digestive system. Our findings indicate that D. melanogaster can serve as a model to further understand EBV-associated gastroinflammatory pathologies. Further studies employing mammalian models may validate the immunogenicity of EBV DNA in an IBD context and its role in exacerbating the disease through inflammatory mediators.
Collapse
Affiliation(s)
- Joelle R Madi
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amani Al Outa
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Mirna Ghannam
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hadi M Hussein
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Marwa Shehab
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Zeinab Al Kobra Haj Hasan
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Antoine Abou Fayad
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Margret Shirinian
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Elias A Rahal
- Department of Experimental Pathology and Immunology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
34
|
Emami-Khoyi A, Knapp IS, Monsanto DM, van Vuuren BJ, Toonen RJ, Teske PR. Genomic divergence and differential gene expression between crustacean ecotypes across a marine thermal gradient. Mar Genomics 2021; 58:100847. [PMID: 33637426 DOI: 10.1016/j.margen.2021.100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/15/2020] [Accepted: 01/29/2021] [Indexed: 11/30/2022]
Abstract
Environmental gradients between marine biogeographical provinces separate distinct faunal communities. However, the absence of absolute dispersal barriers allows numerous species to occur on both sides of such boundaries. While the regional populations of such widespread species are often morphologically indistinguishable from each other, genetic evidence suggests that they represent unique ecotypes, and likely even cryptic species, that may be uniquely adapted to their local environment. Here, we explored genomic divergence in four sympatric southern African decapod crustaceans whose ranges span the boundary between the cool-temperate west coast (south-eastern Atlantic) and the warm-temperate south coast (south-western Indian Ocean) near the southern tip of the African continent. Using genome-wide data, we found that all four species comprise distinct west coast and south coast ecotypes, with molecular dating suggesting divergence during the Pleistocene. Transcriptomic data from the hepatopancreas of twelve specimens of one of these species, the mudprawn Upogebia africana, which were exposed to either 10 °C or 20 °C, showed a clear difference in gene expression profiles between the west- and south coast ecotypes. This difference was particularly clear at 10 °C, where individuals from the south coast experienced a 'transcriptomic shock'. This low temperature is more typical of the west coast during upwelling events, and the physiological stress experienced by the south coast ecotype under such conditions may explain its absence from that coastline. Our results shed new light on the processes involved in driving genomic divergence and incipient speciation along coastlines with porous dispersal barriers.
Collapse
Affiliation(s)
- Arsalan Emami-Khoyi
- Centre for Ecological Genomics and Wildlife Conservation, Department of Zoology, University of Johannesburg, Auckland Park 2006, South Africa.
| | - Ingrid S Knapp
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, Hawai'i, Honolulu, HI, USA
| | - Daniela M Monsanto
- Centre for Ecological Genomics and Wildlife Conservation, Department of Zoology, University of Johannesburg, Auckland Park 2006, South Africa
| | - Bettine Jansen van Vuuren
- Centre for Ecological Genomics and Wildlife Conservation, Department of Zoology, University of Johannesburg, Auckland Park 2006, South Africa
| | - Robert J Toonen
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, Hawai'i, Honolulu, HI, USA
| | - Peter R Teske
- Centre for Ecological Genomics and Wildlife Conservation, Department of Zoology, University of Johannesburg, Auckland Park 2006, South Africa.
| |
Collapse
|
35
|
Schmidt K, Engel P. Mechanisms underlying gut microbiota-host interactions in insects. J Exp Biol 2021; 224:224/2/jeb207696. [PMID: 33509844 DOI: 10.1242/jeb.207696] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Insects are the most diverse group of animals and colonize almost all environments on our planet. This diversity is reflected in the structure and function of the microbial communities inhabiting the insect digestive system. As in mammals, the gut microbiota of insects can have important symbiotic functions, complementing host nutrition, facilitating dietary breakdown or providing protection against pathogens. There is an increasing number of insect models that are experimentally tractable, facilitating mechanistic studies of gut microbiota-host interactions. In this Review, we will summarize recent findings that have advanced our understanding of the molecular mechanisms underlying the symbiosis between insects and their gut microbiota. We will open the article with a general introduction to the insect gut microbiota and then turn towards the discussion of particular mechanisms and molecular processes governing the colonization of the insect gut environment as well as the diverse beneficial roles mediated by the gut microbiota. The Review highlights that, although the gut microbiota of insects is an active field of research with implications for fundamental and applied science, we are still in an early stage of understanding molecular mechanisms. However, the expanding capability to culture microbiomes and to manipulate microbe-host interactions in insects promises new molecular insights from diverse symbioses.
Collapse
Affiliation(s)
- Konstantin Schmidt
- Department of Fundamental Microbiology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Philipp Engel
- Department of Fundamental Microbiology, University of Lausanne, 1015, Lausanne, Switzerland
| |
Collapse
|
36
|
Nagai H, Tatara H, Tanaka-Furuhashi K, Kurata S, Yano T. Homeostatic Regulation of ROS-Triggered Hippo-Yki Pathway via Autophagic Clearance of Ref(2)P/p62 in the Drosophila Intestine. Dev Cell 2021; 56:81-94.e10. [PMID: 33400912 DOI: 10.1016/j.devcel.2020.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/23/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022]
Abstract
Homeostasis of intestinal epithelia is maintained by coordination of the proper rate of regeneration by stem cell division with the rate of cell loss. Regeneration of host epithelia is normally quiescent upon colonization of commensal bacteria; however, the epithelia often develop dysplasia in a context-dependent manner, the cause and underlying mechanism of which remain unclear. Here, we show that in Drosophila intestine, autophagy lowers the sensitivity of differentiated enterocytes to reactive oxygen species (ROS) that are produced in response to commensal bacteria. We find that autophagy deficiency provokes ROS-dependent excessive regeneration and subsequent epithelial dysplasia and barrier dysfunction. Mechanistically, autophagic substrate Ref(2)P/p62, which co-localizes and physically interacts with Dachs, a Hippo signaling regulator, accumulates upon autophagy deficiency and thus inactivates Hippo signaling, resulting in stem cell over-proliferation non-cell autonomously. Our findings uncover a mechanism whereby suppression of undesirable regeneration by autophagy maintains long-term homeostasis of intestinal epithelia.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hiroshi Tatara
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | | | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Tamaki Yano
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
37
|
Satapathy P, Prakash JK, More SS, Chandramohan V, Zameer F. Structural modulation of dual oxidase (Duox) in Drosophila melanogaster by phyto-elicitors: A free energy study with molecular dynamics approach. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
38
|
Yang HT, Huang YH, Yang GW. Mini review: immunologic functions of dual oxidases in mucosal systems of vertebrates. BRAZ J BIOL 2020; 80:948-956. [DOI: 10.1590/1519-6984.208749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 05/08/2019] [Indexed: 12/30/2022] Open
Abstract
Abstract Mucosal epithelial cells act as the first immunologic barrier of organisms, and contact directly with pathogens. Therefore, hosts must have differential strategies to combat pathogens efficiently. Reactive oxygen species (ROS), as a kind of oxidizing agents, participates in the early stage of killing pathogens quickly. Recent reports have revealed that dual oxidase (DUOX) plays a key role in mucosal immunity. And the DUOX is a transmembrane protein which produces ROS as their primary enzymatic products. This process is an important pattern for eliminating pathogens. In this review, we highlight the DUOX immunologic functions in the respiratory and digestive tract of vertebrates.
Collapse
|
39
|
Grenier T, Leulier F. How commensal microbes shape the physiology of Drosophila melanogaster. CURRENT OPINION IN INSECT SCIENCE 2020; 41:92-99. [PMID: 32836177 DOI: 10.1016/j.cois.2020.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023]
Abstract
The interactions between animals and their commensal microbes profoundly influence the host's physiology. In the last decade, Drosophila melanogaster has been extensively used as a model to study host-commensal microbes interactions. Here, we review the most recent advances in this field. We focus on studies that extend our understanding of the molecular mechanisms underlying the effects of commensal microbes on Drosophila's development and lifespan. We emphasize how commensal microbes influence nutrition and the intestinal epithelium homeostasis; how they elicit immune tolerance mechanisms and how these physiological processes are interconnected. Finally, we discuss the importance of diets and microbial strains and show how they can be confounding factors of microbe mediated host phenotypes.
Collapse
Affiliation(s)
- Theodore Grenier
- Univ Lyon, Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, 46, allée d'Italie, 69007, Lyon, France
| | - François Leulier
- Univ Lyon, Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, 46, allée d'Italie, 69007, Lyon, France.
| |
Collapse
|
40
|
Zhou X, Ding G, Li J, Xiang X, Rushworth E, Song W. Physiological and Pathological Regulation of Peripheral Metabolism by Gut-Peptide Hormones in Drosophila. Front Physiol 2020; 11:577717. [PMID: 33117196 PMCID: PMC7552570 DOI: 10.3389/fphys.2020.577717] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
The gastrointestinal (GI) tract in both vertebrates and invertebrates is now recognized as a major source of signals modulating, via gut-peptide hormones, the metabolic activities of peripheral organs, and carbo-lipid balance. Key advances in the understanding of metabolic functions of gut-peptide hormones and their mediated interorgan communication have been made using Drosophila as a model organism, given its powerful genetic tools and conserved metabolic regulation. Here, we summarize recent studies exploring peptide hormones that are involved in the communication between the midgut and other peripheral organs/tissues during feeding conditions. We also highlight the emerging impacts of fly gut-peptide hormones on stress sensing and carbo-lipid metabolism in various disease models, such as energy overload, pathogen infection, and tumor progression. Due to the functional similarity of intestine and its derived peptide hormones between Drosophila and mammals, it can be anticipated that findings obtained in the fly system will have important implications for the understanding of human physiology and pathology.
Collapse
Affiliation(s)
- Xiaoya Zhou
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Guangming Ding
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jiaying Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xiaoxiang Xiang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Elisabeth Rushworth
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
41
|
An Z, Zhao Z, Zhao L, Yue Q, Li K, Zhao B, Miao J, Su L. The novel HOCl fluorescent probe CAN induced A549 apoptosis by inhibiting chlorination activity of MPO. Bioorg Med Chem Lett 2020; 30:127394. [PMID: 32717611 DOI: 10.1016/j.bmcl.2020.127394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 11/18/2022]
Abstract
Hypochlorous acid (HOCl) is an important signaling molecule for cell survival. However, it has been reported that excessive HOCl contributes to a variety of diseases such as cancers. And in cancer cells, the level of HOCl is much higher than that in normal cells. Here a coumarin-based fluorescent probe 7-Diethylamino-3-(2,3-dihydro-1H-perimidin-2-yl)-chromen-2-one (CAN) was successfully developed for HOCl detection. The probe could be oxidized by HOCl to induce significant change in its fluorescence profile, which made it feasible for ratiometric detecting HOCl. CAN (below 1 µM) did not affect cell viability and had good capacity in ratiometric detection of HOCl in RAW 264.7 cells. CAN induced A549 apoptosis and inhibited tumor growth in vitro and in vivo. And CAN could decrease the chlorination activity of myeloperoxidase (MPO) in A549. These findings suggested that CAN (below 1 µM) would develop into a HOCl probe. High activity of MPO and level of HOCl might be helpful for A549 survival. A549 could be induced apoptosis by reducing the HOCl level by CAN. It implies a new anticancer strategy by targeting HOCl.
Collapse
Affiliation(s)
- Zaiyong An
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Zhimin Zhao
- Institute of Developmental Biology, School of Life Science, Shandong University, Qingdao 266237, China
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Qiulin Yue
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Kunlun Li
- Jinan Hangchen Biotechnology Co., Ltd., Jinan 250353, China
| | - Baoxiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Junying Miao
- Institute of Developmental Biology, School of Life Science, Shandong University, Qingdao 266237, China
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| |
Collapse
|
42
|
Ornatowski W, Lu Q, Yegambaram M, Garcia AE, Zemskov EA, Maltepe E, Fineman JR, Wang T, Black SM. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol 2020; 36:101679. [PMID: 32818797 PMCID: PMC7451718 DOI: 10.1016/j.redox.2020.101679] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022] Open
Abstract
The autophagic pathway involves the encapsulation of substrates in double-membraned vesicles, which are subsequently delivered to the lysosome for enzymatic degradation and recycling of metabolic precursors. Autophagy is a major cellular defense against oxidative stress, or related conditions that cause accumulation of damaged proteins or organelles. Selective forms of autophagy can maintain organelle populations or remove aggregated proteins. Dysregulation of redox homeostasis under pathological conditions results in excessive generation of reactive oxygen species (ROS), leading to oxidative stress and the associated oxidative damage of cellular components. Accumulating evidence indicates that autophagy is necessary to maintain redox homeostasis. ROS activates autophagy, which facilitates cellular adaptation and diminishes oxidative damage by degrading and recycling intracellular damaged macromolecules and dysfunctional organelles. The cellular responses triggered by oxidative stress include the altered regulation of signaling pathways that culminate in the regulation of autophagy. Current research suggests a central role for autophagy as a mammalian oxidative stress response and its interrelationship to other stress defense systems. Altered autophagy phenotypes have been observed in lung diseases such as chronic obstructive lung disease, acute lung injury, cystic fibrosis, idiopathic pulmonary fibrosis, and pulmonary arterial hypertension, and asthma. Understanding the mechanisms by which ROS regulate autophagy will provide novel therapeutic targets for lung diseases. This review highlights our current understanding on the interplay between ROS and autophagy in the development of pulmonary disease.
Collapse
Affiliation(s)
- Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Alejandro E Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Emin Maltepe
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
43
|
The Tripartite Interaction of Host Immunity- Bacillus thuringiensis Infection-Gut Microbiota. Toxins (Basel) 2020; 12:toxins12080514. [PMID: 32806491 PMCID: PMC7472377 DOI: 10.3390/toxins12080514] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Bacillus thuringiensis (Bt) is an important cosmopolitan bacterial entomopathogen, which produces various protein toxins that have been expressed in transgenic crops. The evolved molecular interaction between the insect immune system and gut microbiota is changed during the Bt infection process. The host immune response, such as the expression of induced antimicrobial peptides (AMPs), the melanization response, and the production of reactive oxygen species (ROS), varies with different doses of Bt infection. Moreover, B. thuringiensis infection changes the abundance and structural composition of the intestinal bacteria community. The activated immune response, together with dysbiosis of the gut microbiota, also has an important effect on Bt pathogenicity and insect resistance to Bt. In this review, we attempt to clarify this tripartite interaction of host immunity, Bt infection, and gut microbiota, especially the important role of key immune regulators and symbiotic bacteria in the Bt killing activity. Increasing the effectiveness of biocontrol agents by interfering with insect resistance and controlling symbiotic bacteria can be important steps for the successful application of microbial biopesticides.
Collapse
|
44
|
JNK-dependent intestinal barrier failure disrupts host-microbe homeostasis during tumorigenesis. Proc Natl Acad Sci U S A 2020; 117:9401-9412. [PMID: 32277031 PMCID: PMC7196803 DOI: 10.1073/pnas.1913976117] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The intestinal epithelium forms a tight barrier to the environment and is constantly regenerated. Precise control of barrier function and tissue renewal is important to maintain homeostasis. Using an inducible tumor model in the Drosophila intestine, this study shows that tumor progression disrupts the intestinal barrier and leads to commensal dysbiosis, thereby further fueling tumor growth. This reenforcing feedback loop can be interrupted by treatments with JNK inhibitor or antibiotics. In all animals, the intestinal epithelium forms a tight barrier to the environment. The epithelium regulates the absorption of nutrients, mounts immune responses, and prevents systemic infections. Here, we investigate the consequences of tumorigenesis on the microbiome using a Drosophila intestinal tumor model. We show that upon loss of BMP signaling, tumors lead to aberrant activation of JNK/Mmp2 signaling, followed by intestinal barrier dysfunction and commensal imbalance. In turn, the dysbiotic microbiome triggers a regenerative response and stimulates tumor growth. We find that inhibiting JNK signaling or depletion of the microbiome restores barrier function of the intestinal epithelium, leading to a reestablishment of host–microbe homeostasis, and organismic lifespan extension. Our experiments identify a JNK-dependent feedback amplification loop between intestinal tumors and the microbiome. They also highlight the importance of controlling the activity level of JNK signaling to maintain epithelial barrier function and host–microbe homeostasis.
Collapse
|
45
|
Joshi M, Royet J. Uridine Catabolism Breaks the Bonds of Commensalism. Cell Host Microbe 2020; 27:312-314. [PMID: 32164840 DOI: 10.1016/j.chom.2020.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
How gut cells distinguish between beneficial symbionts and deleterious pathobionts is a central question. In this issue of Cell Host & Microbe, Kim et al. (2020) demonstrate that the nucleoside catabolism pathway controlling bacterial uracil and ribose production is an essential trigger of the commensal to pathogen transition.
Collapse
Affiliation(s)
- Manish Joshi
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Julien Royet
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France; Institut Universitaire de France, Paris, France.
| |
Collapse
|
46
|
Bacterial Nucleoside Catabolism Controls Quorum Sensing and Commensal-to-Pathogen Transition in the Drosophila Gut. Cell Host Microbe 2020; 27:345-357.e6. [DOI: 10.1016/j.chom.2020.01.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/03/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023]
|
47
|
Chen J, Aimanova KG, Gill SS. Aedes cadherin receptor that mediates Bacillus thuringiensis Cry11A toxicity is essential for mosquito development. PLoS Negl Trop Dis 2020; 14:e0007948. [PMID: 32012156 PMCID: PMC7018227 DOI: 10.1371/journal.pntd.0007948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 02/13/2020] [Accepted: 11/25/2019] [Indexed: 11/20/2022] Open
Abstract
Aedes cadherin (AaeCad, AAEL024535) has been characterized as a receptor for Bacillus thuringiensis subsp. israelensis (Bti) Cry11A toxins. However, its role in development is still unknown. In this study, we modified the cadherin gene using ZFN and TALEN. Even though we obtained heterozygous deletions, no homozygous mutants were viable. Because ZFN and TALEN have lower off-targets than CRISPR/Cas9, we conclude the cadherin gene is essential for Aedes development. In contrast, in lepidopteran insects loss of a homologous cadherin does not appear to be lethal, since homozygous mutants are viable. To analyze the role of AaeCad in vivo, we tagged this protein with EGFP using CRISPR-Cas9-mediated homologous recombination and obtained a homozygous AaeCad-EGFP line. Addition of Aedes Rad51 mRNA enhanced the rate of recombination. We then examined AaeCad protein expression in most tissues and protein dynamics during mosquito development. We observe that AaeCad is expressed in larval and adult midgut-specific manner and its expression pattern changed during the mosquito development. Confocal images showed AaeCad has high expression in larval caecae and posterior midgut, and also in adult midgut. Expression of AaeCad is observed primarily in the apical membranes of epithelial cells, and not in cell-cell junctions. The expression pattern observed suggests AaeCad does not appear to play a role in these junctions. However, we cannot exclude its role beyond cell-cell adhesion in the midgut. We also observed that Cry11A bound to the apical side of larval gastric caecae and posterior midgut cells exactly where AaeCad-EGFP was expressed. Their co-localization suggests that AaeCad is indeed a receptor for the Cry11A toxin. Using this mosquito line we also observed that low doses of Cry11A toxin caused the cells to slough off membranes, which likely represents a defense mechanism, to limit cell damage from Cry11A toxin pores formed in the cell membrane. A number of receptors for Bt Cry toxins, have been identified and characterized, including cadherin proteins. However, the role of these proteins in the insect is unknown and there have been few efforts to elucidate their function. First, in this study we show that in the mosquito, Aedes aegypti, the cadherin protein is essential for development. Secondly, we provide evidence that AaeCad plays a role in the apical membrane and the maintenance of midgut integrity by gene tagging using CRISPR/Cas9, which overcomes the limitation of receptor localization using antibodies in previous studies. These investigations are helpful to further investigate the physiological function of AaeCad. Moreover, this study demonstrated successful tagging of an essential gene with fluorescence protein in a non-model insect. In addition, this study showed that epithelium thinning is possibly a conserved mechanism for host defense against pore-forming toxins, like Cry11A.
Collapse
Affiliation(s)
- Jianwu Chen
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, United States of America
| | - Karly G. Aimanova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, United States of America
| | - Sarjeet S. Gill
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Tarashi S, Siadat SD, Ahmadi Badi S, Zali M, Biassoni R, Ponzoni M, Moshiri A. Gut Bacteria and their Metabolites: Which One Is the Defendant for Colorectal Cancer? Microorganisms 2019; 7:E561. [PMID: 31766208 PMCID: PMC6920974 DOI: 10.3390/microorganisms7110561] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/22/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a worldwide health concern which requires efficient therapeutic strategies. The mechanisms underlying CRC remain an essential subject of investigations in the cancer biology field. The evaluation of human microbiota can be critical in this regard, since the disruption of the normal community of gut bacteria is an important issue in the development of CRC. However, several studies have already evaluated the different aspects of the association between microbiota and CRC. The current study aimed at reviewing and summarizing most of the studies on the modifications of gut bacteria detected in stool and tissue samples of CRC cases. In addition, the importance of metabolites derived from gut bacteria, their relationship with the microbiota, and epigenetic modifications have been evaluated.
Collapse
Affiliation(s)
- Samira Tarashi
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Sara Ahmadi Badi
- Microbiology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran; (S.T.); (S.D.S.); (S.A.B.)
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Mohammadreza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, 19857-17411 Tehran, Iran;
| | - Roberto Biassoni
- Laboratory of Molecular Medicine, IRCCS Instituto Giannina Gaslini, 16147 Genova, Italy;
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Arfa Moshiri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, 19857-17411 Tehran, Iran;
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| |
Collapse
|
49
|
An infection of Enterobacter ludwigii affects development and causes age-dependent neurodegeneration in Drosophila melanogaster. INVERTEBRATE NEUROSCIENCE 2019; 19:13. [PMID: 31641932 DOI: 10.1007/s10158-019-0233-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023]
Abstract
The effects of teeth-blackening bacteria Enterobacter ludwigii on the physiological system were investigated using the model organism Drosophila melanogaster. The bacteria were mixed with the fly food, and its effect was checked on the growth, development and behaviour of Drosophila. Microbes generate reactive oxygen species (ROS) within the haemolymph of the larvae once it enters into the body. The increased amount of ROS was evidenced by the NBT assay and using 2',7'-dichlorofluorescin diacetate dye, which indicates the mitochondrial ROS. The increased amount of ROS resulted in a number of abnormal nuclei within the gut. Besides that larvae walking became sluggish in comparison with wild type although the larvae crawling path did not change much. Flies hatched from the infectious larvae have the posterior scutellar bristle absent from the thorax and abnormal mechanosensory hairs in the eye, and they undergo time-dependent neurodegeneration as evidenced by the geotrophic and phototrophic assays. To decipher the mechanism of neurodegeneration, flies were checked for the presence of four important bioamines: tyramine, cadaverine, putrescine and histamine. Out of these four, histamine was found to be absent in infected flies. Histamine is a key molecule required for the functioning of the photoreceptor as well as mechanoreceptors. The mechanism via which mouth infectious bacteria E. ludwigii can affect the development and cause age-dependent neurodegeneration is explained in this paper.
Collapse
|
50
|
Abstract
Sublethal exposure to certain pesticides (e.g., neonicotinoid insecticides) is suspected to contribute to honey bee (Apis mellifera) population decline in North America. Neonicotinoids are known to interfere with immune pathways in the gut of insects, but the underlying mechanisms remain elusive. We used a Drosophila melanogaster model to understand how imidacloprid (a common neonicotinoid) interferes with two innate immune pathways—Duox and Imd. We found that imidacloprid dysregulates these pathways to reduce hydrogen peroxide production, ultimately leading to a dysbiotic shift in the gut microbiota. Intriguingly, we found that presupplementation with probiotic bacteria could mitigate the harmful effects of imidacloprid. Thus, these observations uncover a novel mechanism of pesticide-induced immunosuppression that exploits the interconnectedness of two important insect immune pathways. Neonicotinoid insecticides are common agrochemicals that are used to kill pest insects and improve crop yield. However, sublethal exposure can exert unintentional toxicity to honey bees and other beneficial pollinators by dysregulating innate immunity. Generation of hydrogen peroxide (H2O2) by the dual oxidase (Duox) pathway is a critical component of the innate immune response, which functions to impede infection and maintain homeostatic regulation of the gut microbiota. Despite the importance of this pathway in gut immunity, the consequences of neonicotinoid exposure on Duox signaling have yet to be studied. Here, we use a Drosophila melanogaster model to investigate the hypothesis that imidacloprid (a common neonicotinoid) can affect the Duox pathway. The results demonstrated that exposure to sublethal imidacloprid reduced H2O2 production by inhibiting transcription of the Duox gene. Furthermore, the reduction in Duox expression was found to be a result of imidacloprid interacting with the midgut portion of the immune deficiency pathway. This impairment led to a loss of microbial regulation, as exemplified by a compositional shift and increased total abundance of Lactobacillus and Acetobacter spp. (dominant microbiota members) found in the gut. In addition, we demonstrated that certain probiotic lactobacilli could ameliorate Duox pathway impairment caused by imidacloprid, but this effect was not directly dependent on the Duox pathway itself. This study is the first to demonstrate the deleterious effects that neonicotinoids can have on Duox-mediated generation of H2O2 and highlights a novel coordination between two important innate immune pathways present in insects.
Collapse
|