1
|
Zhang H, Zhu Y, Wang Y, Jiang L, Shi X, Cheng G. Microbial interactions shaping host attractiveness: insights into dynamic behavioral relationships. CURRENT OPINION IN INSECT SCIENCE 2024; 66:101275. [PMID: 39332621 DOI: 10.1016/j.cois.2024.101275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 07/18/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
Insects discern the presence of hosts (host plants) by integrating chemosensory, gustatory, and visual cues, with olfaction playing a pivotal role in this process. Among these factors, volatile signals produced by host-associated microbial communities significantly affect insect attraction. Microorganisms are widely and abundantly found on the surfaces of humans, plants, and insects. Notably, these microorganisms can metabolize compounds from the host surface and regulate the production of characteristic volatiles, which may guide the use of host microorganisms to modulate insect behavior. Essentially, the attraction of hosts to insects is intricately linked to the presence of their symbiotic microorganisms. This review underscores the critical role of microorganisms in shaping the dynamics of attractiveness between insects and their hosts.
Collapse
Affiliation(s)
- Hong Zhang
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yibin Zhu
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Southwest United Graduate School, Kunming 650092, China
| | - Yibaina Wang
- China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Liping Jiang
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Xiaolu Shi
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Southwest United Graduate School, Kunming 650092, China.
| |
Collapse
|
2
|
Sousa M, Magalhães R, Ferreira V, Teixeira P. Current methodologies available to evaluate the virulence potential among Listeria monocytogenes clonal complexes. Front Microbiol 2024; 15:1425437. [PMID: 39493856 PMCID: PMC11528214 DOI: 10.3389/fmicb.2024.1425437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis in humans, the severity of which depends on multiple factors, including intrinsic characteristics of the affected individuals and the pathogen itself. Additionally, emerging evidence suggests that epigenetic modifications may also modulate host susceptibility to infection. Therefore, different clinical outcomes can be expected, ranging from self-limiting gastroenteritis to severe central nervous system and maternal-neonatal infections, and bacteremia. Furthermore, L. monocytogenes is a genetically and phenotypically diverse species, resulting in a large variation in virulence potential between strains. Multilocus sequence typing (MLST) has been widely used to categorize the clonal structure of bacterial species and to define clonal complexes (CCs) of genetically related isolates. The combination of MLST and epidemiological data allows to distinguish hypervirulent CCs, which are notably more prevalent in clinical cases and typically associated with severe forms of the disease. Conversely, other CCs, termed hypovirulent, are predominantly isolated from food and food processing environments and are associated with the occurrence of listeriosis in immunosuppressed individuals. Reports of genetic traits associated with this diversity have been described. The Food and Agriculture Organization (FAO) is encouraging the search for virulence biomarkers to rapidly identify the main strains of concern to reduce food waste and economical losses. The aim of this review is to comprehensively collect, describe and discuss the methodologies used to discriminate the virulence potential of L. monocytogenes CCs. From the exploration of in vitro and in vivo models to the study of expression of virulence genes, each approach is critically explored to better understand its applicability and efficiency in distinguishing the virulence potential of the pathogen.
Collapse
Affiliation(s)
| | | | | | - Paula Teixeira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, Portugal
| |
Collapse
|
3
|
Dos Santos Moreira LM, Marinho LS, Neves RCS, Harakava R, Bessa LA, Vitorino LC. Assessment of the Entomopathogenic Potential of Fungal and Bacterial Isolates from Fall Armyworm Cadavers Against Spodoptera frugiperda Caterpillars and the Adult Boll Weevil, Anthonomus grandis. NEOTROPICAL ENTOMOLOGY 2024; 53:889-906. [PMID: 38714593 PMCID: PMC11255027 DOI: 10.1007/s13744-024-01159-0] [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: 11/20/2023] [Accepted: 04/13/2024] [Indexed: 05/10/2024]
Abstract
Increased attention is being focused on the biological control of agricultural pests using microorganisms, owing to their potential as a viable substitute for chemical control methods. Insect cadavers constitute a potential source of entomopathogenic microorganisms. We tested whether bacteria and fungi isolated from Spodoptera frugiperda (JE Smith) cadavers could affect its survival, development, egg-laying pattern, and hatchability, as well as induce mortality in Anthonomus grandis Boheman adults. We isolated the bacteria Enterobacter hormaechei and Serratia marcescens and the fungi Scopulariopsis sp. and Aspergillus nomiae from fall armyworm cadavers and the pest insects were subjected to an artificial diet enriched with bacteria cells or fungal spores to be tested, in the case of S. frugiperda, and only fungal spores in the case of A. grandis. Enterobacter hormaechei and A. nomiae were pathogenic to S. frugiperda, affecting the survival of adults and pupae. The fungus Scopulariopsis sp. does not affect the survival of S. frugiperda caterpillars and pupae; however, due to late action, moths and eggs may be affected. Aspergillus nomiae also increased mortality of A. grandis adults, as well as the development of S. frugiperda in the early stages of exposure to the diet, as indicated by the vertical spore transfer to offspring and low hatchability. Enterobacter hormaechei and A. nomiae are potential biocontrol agents for these pests, and warrant further investigation from a toxicological point of view and subsequently in field tests involving formulations that could improve agricultural sustainability practices.
Collapse
Affiliation(s)
- Lidiane Maria Dos Santos Moreira
- Instituto Goiano de Agricultura (IGA), Montividiu, GO, Brazil
- Lab of Agricultural Microbiology, Instituto Federal Goiano, Rio Verde Campus, Rio Verde, GO, Brazil
| | | | | | | | - Layara Alexandre Bessa
- Lab of Biodiversity Metabolism and Genetics, Instituto Federal Goiano, Rio Verde Campus, Rio Verde, GO, Brazil
- Simple Agro Corporation, Rio Verde, GO, Brazil
| | - Luciana Cristina Vitorino
- Simple Agro Corporation, Rio Verde, GO, Brazil.
- Lab of Agricultural Microbiology, Instituto Federal Goiano, Rio Verde Campus, Rio Verde, GO, Brazil.
| |
Collapse
|
4
|
Darby AM, Okoro DO, Aredas S, Frank AM, Pearson WH, Dionne MS, Lazzaro BP. High sugar diets can increase susceptibility to bacterial infection in Drosophila melanogaster. PLoS Pathog 2024; 20:e1012447. [PMID: 39133760 PMCID: PMC11341100 DOI: 10.1371/journal.ppat.1012447] [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: 12/17/2023] [Revised: 08/22/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Overnutrition with dietary sugar can worsen infection outcomes in diverse organisms including insects and humans, through generally unknown mechanisms. In the present study, we show that adult Drosophila melanogaster fed high-sugar diets became more susceptible to infection by the Gram-negative bacteria Providencia rettgeri and Serratia marcescens. We found that P. rettgeri and S. marcescens proliferate more rapidly in D. melanogaster fed a high-sugar diet, resulting in increased probability of host death. D. melanogaster become hyperglycemic on the high-sugar diet, and we find evidence that the extra carbon availability may promote S. marcescens growth within the host. However, we found no evidence that increased carbon availability directly supports greater P. rettgeri growth. D. melanogaster on both diets fully induce transcription of antimicrobial peptide (AMP) genes in response to infection, but D. melanogaster provided with high-sugar diets show reduced production of AMP protein. Thus, overnutrition with dietary sugar may impair host immunity at the level of AMP translation. Our results demonstrate that dietary sugar can shape infection dynamics by impacting both host and pathogen, depending on the nutritional requirements of the pathogen and by altering the physiological capacity of the host to sustain an immune response.
Collapse
Affiliation(s)
- Andrea M. Darby
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| | - Destiny O. Okoro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| | - Sophia Aredas
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
- University of California, Irvine, Irvine, California, United States of America
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - Ashley M. Frank
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Battelle, Columbus, Ohio, United States of America
| | - William H. Pearson
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Marc S. Dionne
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
5
|
Moyano A, Croce AC, Scolari F. Pathogen-Mediated Alterations of Insect Chemical Communication: From Pheromones to Behavior. Pathogens 2023; 12:1350. [PMID: 38003813 PMCID: PMC10675518 DOI: 10.3390/pathogens12111350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Pathogens can influence the physiology and behavior of both animal and plant hosts in a manner that promotes their own transmission and dispersal. Recent research focusing on insects has revealed that these manipulations can extend to the production of pheromones, which are pivotal in chemical communication. This review provides an overview of the current state of research and available data concerning the impacts of bacterial, viral, fungal, and eukaryotic pathogens on chemical communication across different insect orders. While our understanding of the influence of pathogenic bacteria on host chemical profiles is still limited, viral infections have been shown to induce behavioral changes in the host, such as altered pheromone production, olfaction, and locomotion. Entomopathogenic fungi affect host chemical communication by manipulating cuticular hydrocarbons and pheromone production, while various eukaryotic parasites have been observed to influence insect behavior by affecting the production of pheromones and other chemical cues. The effects induced by these infections are explored in the context of the evolutionary advantages they confer to the pathogen. The molecular mechanisms governing the observed pathogen-mediated behavioral changes, as well as the dynamic and mutually influential relationships between the pathogen and its host, are still poorly understood. A deeper comprehension of these mechanisms will prove invaluable in identifying novel targets in the perspective of practical applications aimed at controlling detrimental insect species.
Collapse
Affiliation(s)
- Andrea Moyano
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| | - Anna Cleta Croce
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| | - Francesca Scolari
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| |
Collapse
|
6
|
Darby AM, Lazzaro BP. Interactions between innate immunity and insulin signaling affect resistance to infection in insects. Front Immunol 2023; 14:1276357. [PMID: 37915572 PMCID: PMC10616485 DOI: 10.3389/fimmu.2023.1276357] [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: 08/11/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023] Open
Abstract
An active immune response is energetically demanding and requires reallocation of nutrients to support resistance to and tolerance of infection. Insulin signaling is a critical global regulator of metabolism and whole-body homeostasis in response to nutrient availability and energetic needs, including those required for mobilization of energy in support of the immune system. In this review, we share findings that demonstrate interactions between innate immune activity and insulin signaling primarily in the insect model Drosophila melanogaster as well as other insects like Bombyx mori and Anopheles mosquitos. These studies indicate that insulin signaling and innate immune activation have reciprocal effects on each other, but that those effects vary depending on the type of pathogen, route of infection, and nutritional status of the host. Future research will be required to further understand the detailed mechanisms by which innate immunity and insulin signaling activity impact each other.
Collapse
Affiliation(s)
- Andrea M. Darby
- Department of Entomology, Cornell University, Ithaca, NY, United States
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, United States
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, NY, United States
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, United States
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
Andrade LC, Majerowicz D, Oliveira PL, Guarneri AA. Alterations in energy metabolism of Rhodnius prolixus induced by Trypanosoma rangeli infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 159:103987. [PMID: 37429385 DOI: 10.1016/j.ibmb.2023.103987] [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: 03/24/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
Trypanosoma rangeli is a protozoan parasite that infects triatomines and mammals in the Americas, producing mixed infections with Trypanosoma cruzi, the etiological agent of Chagas disease. The former parasite is not pathogenic to humans, but has different levels of pathogenicity, as well as causing physiological and behavioral alterations, to its invertebrate hosts. In this study, we measured locomotory activity, and the glyceride accumulation profile in the hemolymph and fat body, as well as the expression of key genes related to triglyceride metabolism, of Rhodnius prolixus nymphs infected with T. rangeli. We found that the locomotory activity of the insects was correlated with the amount of triglycerides in the fat body. Infected nymphs had increased activity when starved, and also had an accumulation of glycerides in the fat body and hemolymph. These alterations were also associated with a higher expression of the diacylglycerol acyltransferase, lipophorin and lipophorin receptor genes in the fat body. We infer that T. rangeli is able to alter the energetic processes of its invertebrate host, in order to increase the availability of lipids to the parasite, which, in turn modifies the activity levels of the insect. These alterations are discussed with regard to their potential to increase the transmission rate of the parasite.
Collapse
Affiliation(s)
- Laila C Andrade
- Vector Behavior and Pathogen Interaction Group, Instituto René Rachou, Fiocruz, Avenida Augusto de Lima, 1715, Belo Horizonte, MG, CEP 30190-009, Brazil
| | - David Majerowicz
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Prédio do CCS, bloco A, 2° andar, sala 48, CEP: 21941-590, Cidade Universitária, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Biociências, Universidade do Estado do Rio de Janeiro, Brazil; INCT-EM, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Ilha do Fundão, Rio de Janeiro, 21941-902, Brazil; INCT-EM, Brazil
| | - Alessandra A Guarneri
- Vector Behavior and Pathogen Interaction Group, Instituto René Rachou, Fiocruz, Avenida Augusto de Lima, 1715, Belo Horizonte, MG, CEP 30190-009, Brazil; INCT-EM, Brazil.
| |
Collapse
|
9
|
Ozakman Y, Raval D, Eleftherianos I. Drosophila melanogaster Imd signaling interacts with insulin signaling and alters feeding rate upon parasitic nematode infection. Heliyon 2023; 9:e16139. [PMID: 37251825 PMCID: PMC10208921 DOI: 10.1016/j.heliyon.2023.e16139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/15/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Significant progress has been made in recent years on exploring immunometabolism, a field that integrates two processes essential for maintaining tissue and organismal homeostasis, immunity and metabolism. The nematode parasite Heterorhabditis gerrardi, its mutualistic bacteria Photorhabdus asymbiotica, and the fruit fly Drosophila melanogaster constitute a unique system to investigate the molecular basis of host immunometabolic response to nematode-bacterial complexes. In this study, we explored the contribution of the two major immune signaling pathways, Toll and Imd, to sugar metabolism in D. melanogaster larvae during infection with H. gerrardi nematodes. We infected Toll or Imd signaling loss-of-function mutant larvae with H. gerrardi nematodes and assessed larval survival ability, feeding rate, and sugar metabolism. We found no significant differences in the survival ability or levels of sugar metabolites in any of the mutant larvae when responding to H. gerrardi infection. However, we found that the Imd mutant larvae have higher feeding rate than controls during the early stages of infection. In addition, feeding rates are lower in Imd mutants relative to the control larvae as the infection progresses. We further showed that Dilp2 and Dilp3 gene expression increases in Imd mutants compared to controls early in the infection, but their expression levels decrease at later times. These findings indicate that Imd signaling activity regulates the feeding rate and Dilp2 and Dilp3 expression in D. melanogaster larvae infected with H. gerrardi. Results from this study facilitate our understanding of the link between host innate immunity and sugar metabolism in the context of infectious diseases caused by parasitic nematodes.
Collapse
Affiliation(s)
- Yaprak Ozakman
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington DC, 20052, USA
| | - Dhaivat Raval
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington DC, 20052, USA
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington DC, 20052, USA
| |
Collapse
|
10
|
Zhang L, Parreira VR, Rahman A, Smith BA, Munther DS, Farber JM. Survival and predictive modeling of Listeria monocytogenes under simulated human gastric conditions in the presence of bovine milk products. Int J Food Microbiol 2023; 396:110201. [PMID: 37116301 DOI: 10.1016/j.ijfoodmicro.2023.110201] [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: 06/16/2022] [Revised: 03/15/2023] [Accepted: 04/01/2023] [Indexed: 04/30/2023]
Abstract
Listeria monocytogenes is an opportunistic foodborne pathogen which has been implicated in many outbreaks of foodborne diseases. This study evaluated the effects of gastric acidity and gastric digestion time of adults, L. monocytogenes strain and food type on the survival of L. monocytogenes under simulated stomach conditions of adults in in vitro gastric models with dynamic pH changes occurring throughout the exposure. Individual strains as well as a cocktail of L. monocytogenes, inoculated at 8 log CFU/mL in filtered bovine milk products, 0 % milk, 2 % milk, 2 % chocolate milk and 3.25 % milk, were introduced to the gastric models for 2 h. The survival of L. monocytogenes depended on a combination of factors, including gastric acidity and gastric digestion time of adults, L. monocytogenes strain, food type and recovery method (P < 0.05). The survival rates of L. monocytogenes inoculated in 2 % milk after a 2-h exposure to simulated gastric fluids with pH values of 1.5, 2.0 and 3.0 were 0.003 to 0.040 %, 22.7 to 43.4 % and 16.6 to 27.2 %, respectively. Fluid milk containing a higher milk fat content (3.25 % vs 0 % milk) protected L. monocytogenes from being inactivated when they were exposed to the human stomach model with a gastric acidity of pH 2.0. Compared to 0 % and 3.25 % milk, L. monocytogenes survived the best in 2 % chocolate milk, which appears to be due to the presence of milk fat (2 %) and the additional nutrients that are present in chocolate milk. A predictive mathematical model was developed that captured the population of the strains of L. monocytogenes under the in vitro conditions. This study advances our understanding of the behaviour of L. monocytogenes under various human gastric conditions and provides key parameters that can affect the survival of L. monocytogenes in the stomachs of adults. The mathematical models developed in this study can be used as a supplementary tool to help predict the survival of L. monocytogenes under similar scenarios and for relevant risk-assessment studies.
Collapse
Affiliation(s)
- Linkang Zhang
- Canadian Research Institute for Food Safety, Department of Food Science, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Valeria R Parreira
- Canadian Research Institute for Food Safety, Department of Food Science, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Ashrafur Rahman
- Department of Mathematics and Statistics, Oakland University, Rochester, MI 48309, USA
| | - Ben A Smith
- National Microbiology Laboratory, Public Health Agency of Canada, Guelph, ON N1H 7M7, Canada
| | - Daniel S Munther
- Department of Mathematics and Statistics, Cleveland State University, Cleveland, OH 44115, USA
| | - Jeffrey M Farber
- Canadian Research Institute for Food Safety, Department of Food Science, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
11
|
Bland ML. Regulating metabolism to shape immune function: Lessons from Drosophila. Semin Cell Dev Biol 2023; 138:128-141. [PMID: 35440411 PMCID: PMC10617008 DOI: 10.1016/j.semcdb.2022.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/21/2022] [Accepted: 04/03/2022] [Indexed: 12/14/2022]
Abstract
Infection with pathogenic microbes is a severe threat that hosts manage by activating the innate immune response. In Drosophila melanogaster, the Toll and Imd signaling pathways are activated by pathogen-associated molecular patterns to initiate cellular and humoral immune processes that neutralize and kill invaders. The Toll and Imd signaling pathways operate in organs such as fat body and gut that control host nutrient metabolism, and infections or genetic activation of Toll and Imd signaling also induce wide-ranging changes in host lipid, carbohydrate and protein metabolism. Metabolic regulation by immune signaling can confer resistance to or tolerance of infection, but it can also lead to pathology and susceptibility to infection. These immunometabolic phenotypes are described in this review, as are changes in endocrine signaling and gene regulation that mediate survival during infection. Future work in the field is anticipated to determine key variables such as sex, dietary nutrients, life stage, and pathogen characteristics that modify immunometabolic phenotypes and, importantly, to uncover the mechanisms used by the immune system to regulate metabolism.
Collapse
Affiliation(s)
- Michelle L Bland
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, United States.
| |
Collapse
|
12
|
Abbas MN, Kausar S, Asma B, Ran W, Li J, Lin Z, Li T, Cui H. MicroRNAs reshape the immunity of insects in response to bacterial infection. Front Immunol 2023; 14:1176966. [PMID: 37153604 PMCID: PMC10161253 DOI: 10.3389/fimmu.2023.1176966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
The interaction between bacteria and insects can significantly impact a wide range of different areas because bacteria and insects are widely distributed around the globe. The bacterial-insect interactions have the potential to directly affect human health since insects are vectors for disease transmission, and their interactions can also have economic consequences. In addition, they have been linked to high mortality rates in economically important insects, resulting in substantial economic losses. MicroRNAs (miRNAs) are types of non-coding RNAs involved in regulating gene expression post-transcriptionally. The length of miRNAs ranges from 19 to 22 nucleotides. MiRNAs, in addition to their ability to exhibit dynamic expression patterns, have a diverse range of targets. This enables them to govern various physiological activities in insects, like innate immune responses. Increasing evidence suggests that miRNAs have a crucial biological role in bacterial infection by influencing immune responses and other mechanisms for resistance. This review focuses on some of the most recent and exciting discoveries made in recent years, including the correlation between the dysregulation of miRNA expression in the context of bacterial infection and the progression of the infection. Furthermore, it describes how they profoundly impact the immune responses of the host by targeting the Toll, IMD, and JNK signaling pathways. It also emphasizes the biological function of miRNAs in regulating immune responses in insects. Finally, it also discusses current knowledge gaps about the function of miRNAs in insect immunity, in addition to areas that require more research in the future.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Saima Kausar
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Bibi Asma
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Wenhao Ran
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Jingui Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Zini Lin
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Tiejun Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
- *Correspondence: Tiejun Li, ; Hongjuan Cui,
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- *Correspondence: Tiejun Li, ; Hongjuan Cui,
| |
Collapse
|
13
|
Infection increases activity via Toll dependent and independent mechanisms in Drosophila melanogaster. PLoS Pathog 2022; 18:e1010826. [PMID: 36129961 PMCID: PMC9529128 DOI: 10.1371/journal.ppat.1010826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 10/03/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Host behavioural changes are among the most apparent effects of infection. ‘Sickness behaviour’ can involve a variety of symptoms, including anorexia, depression, and changed activity levels. Here, using a real-time tracking and behavioural profiling platform, we show that in Drosophila melanogaster, several systemic bacterial infections cause significant increases in physical activity, and that the extent of this activity increase is a predictor of survival time in some lethal infections. Using multiple bacteria and D. melanogaster immune and activity mutants, we show that increased activity is driven by at least two different mechanisms. Increased activity after infection with Micrococcus luteus, a Gram-positive bacterium rapidly cleared by the immune response, strictly requires the Toll ligand spätzle. In contrast, increased activity after infection with Francisella novicida, a Gram-negative bacterium that cannot be cleared by the immune response, is entirely independent of both Toll and the parallel IMD pathway. The existence of multiple signalling mechanisms by which bacterial infections drive increases in physical activity implies that this effect may be an important aspect of the host response. Sickness behaviours are often observed during infection. Animals have been shown to change their feeding, mating, social and resting (sleeping) behaviours in response to infection. We show here that fruit-flies infected with bacteria respond by increasing their physical activity and decreasing the amount of time spent sleeping. This increase in activity is seen in some, but not all, bacterial infections, and appears to be driven by at least two different mechanisms: with some bacteria, activating the immune response is the only requirement to induce increased activity, while other bacteria induce increased activity independently of known immune detection pathways. The biological role of increased activity is unclear; flies in the wild may be driven to flee sites where infection risk or pathogen burden is high. Alternatively, increased activity could serve a less direct anti-microbial function. For example, active animals may be more likely to encounter potential mates or food resource.
Collapse
|
14
|
Luo W, Liu S, Zhang F, Zhao L, Su Y. Metabolic strategy of macrophages under homeostasis or immune stress in Drosophila. MARINE LIFE SCIENCE & TECHNOLOGY 2022; 4:291-302. [PMID: 37073169 PMCID: PMC10077226 DOI: 10.1007/s42995-022-00134-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/06/2022] [Indexed: 05/03/2023]
Abstract
Macrophages are well known for their phagocytic functions in innate immunity across species. In mammals, they rapidly consume a large amount of energy by shifting their metabolism from mitochondrial oxidative phosphorylation toward aerobic glycolysis, to perform the effective bactericidal function upon infection. Meanwhile, they strive for sufficient energy resources by restricting systemic metabolism. In contrast, under nutrient deprivation, the macrophage population is down-regulated to save energy for survival. Drosophila melanogaster possesses a highly conserved and comparatively simple innate immune system. Intriguingly, recent studies have shown that Drosophila plasmatocytes, the macrophage-like blood cells, adopt comparable metabolic remodeling and signaling pathways to achieve energy reassignment when challenged by pathogens, indicating the conservation of such metabolic strategies between insects and mammals. Here, focusing on Drosophila macrophages (plasmatocytes), we review recent advances regarding their comprehensive roles in local or systemic metabolism under homeostasis or stress, emphasizing macrophages as critical players in the crosstalk between the immune system and organic metabolism from a Drosophila perspective.
Collapse
Affiliation(s)
- Wang Luo
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Sumin Liu
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Fang Zhang
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Long Zhao
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- Fisheries College, Ocean University of China, Qingdao, 266003 China
- Key Laboratory of Mariculture (OUC), Ministry of Education, Qingdao, 266003 China
| | - Ying Su
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| |
Collapse
|
15
|
Rose S, Beckwith EJ, Burmester C, May RC, Dionne MS, Rezaval C. Pre-copulatory reproductive behaviours are preserved in Drosophila melanogaster infected with bacteria. Proc Biol Sci 2022; 289:20220492. [PMID: 35538789 PMCID: PMC9091859 DOI: 10.1098/rspb.2022.0492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The activation of the immune system upon infection exerts a huge energetic demand on an individual, likely decreasing available resources for other vital processes, like reproduction. The factors that determine the trade-off between defensive and reproductive traits remain poorly understood. Here, we exploit the experimental tractability of the fruit fly Drosophila melanogaster to systematically assess the impact of immune system activation on pre-copulatory reproductive behaviour. Contrary to expectations, we found that male flies undergoing an immune activation continue to display high levels of courtship and mating success. Similarly, immune-challenged female flies remain highly sexually receptive. By combining behavioural paradigms, a diverse panel of pathogens and genetic strategies to induce the fly immune system, we show that pre-copulatory reproductive behaviours are preserved in infected flies, despite the significant metabolic cost of infection.
Collapse
Affiliation(s)
- Saloni Rose
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Esteban J. Beckwith
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London SW7 2AZ, UK,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), UBA-CONICET, Buenos Aires, Argentina
| | | | - Robin C. May
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK,Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, UK
| | - Marc S. Dionne
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Carolina Rezaval
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
16
|
Wu G, Liu J, Li M, Xiao Y, Yi Y. Prior infection of Galleria mellonella with sublethal dose of Bt elicits immune priming responses but incurs metabolic changes. JOURNAL OF INSECT PHYSIOLOGY 2022; 139:104401. [PMID: 35636486 DOI: 10.1016/j.jinsphys.2022.104401] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Invertebrate immune priming has attracted wide attention of biologists in recent years because it challenges core notions about the disparate nature of acquired and innate immunity. However, the metabolic switch and energetic cost during eliciting immune priming are poorly investigated issues, which could widen and deepen our understanding of the physiological mechanism of immune priming. In this study, using sublethal dose of Bacillus thuringiensis (Bt) as an elicitor, we detected typical immune priming responses in Galleria mellonella. We found that the intensity of immune priming is positively correlated with the levels of antimicrobial peptides and phagocytosis ability of hemocytes. Subsequently, we employed LC-MS/MS-based untargeted metabolomics techniques to analyze the metabolic changes in the fat body of G. mellonella larvae during immune priming. The results showed that there were 74 and 56 significantly altered metabolites in positive and negative ion mode, respectively, after Bt priming. Most of the differential metabolites were enriched in the following metabolic pathways: amino acid biosynthesis, carbon metabolism, aminoacyl-tRNA biosynthesis and ABC transporters. The energetic cost of immune priming was depicted mainly in the slow growth of body mass and decreased levels of sucrose, lactose, D-ribulose 1,5-bisphosphate, Glycerate-3P and isocitric acid, which are enriched in carbon metabolism and involved in energy production. Meanwhile, correlation and interaction network analysis showed negative correlations between carbohydrates and metabolites involved in amino acid biosynthesis, suggesting that amino acids acted as the main energy source and helped the organisms synthesize immune effectors to participate in the immune priming response. Our results pave the way for uncovering the physiological mechanism of insect immune priming and discovering novel targets for Bt insecticide.
Collapse
Affiliation(s)
- Gongqing Wu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China; Guangdong Cosmetics Engineering & Technology Research Center, Zhongshan 528458, PR China
| | - Jiajie Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China
| | - Mei Li
- University of Electronic Science and Technology of China, Zhongshan Institute, Zhongshan 528402, PR China
| | - Yang Xiao
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Science, Guangzhou 510610, PR China
| | - Yunhong Yi
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China.
| |
Collapse
|
17
|
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: 0.7] [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
|
18
|
Application of metabolomics analysis to aid in understanding the pathogenicity of different lineages and different serotypes of Listeria monocytogenes. Int J Food Microbiol 2022; 373:109694. [DOI: 10.1016/j.ijfoodmicro.2022.109694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/19/2022]
|
19
|
Kwon H, Smith R. Anopheles gambiae Actively Metabolizes Uric Acid Following Plasmodium Infection to Limit Malaria Parasite Survival. Front Physiol 2022; 12:821869. [PMID: 35140633 PMCID: PMC8818946 DOI: 10.3389/fphys.2021.821869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Characterizing the physiological changes that accompany malaria parasite infection of the mosquito host is crucial to our understanding of vectorial capacity in Anopheles mosquitoes, yet has not fully been explored. In this study, we examine the role of uric acid metabolism in the mosquito, Anopheles gambiae, following malaria parasite infection. We demonstrate that levels of uric acid are significantly decreased in the excreta and the mosquito at 24 and 48 h post-Plasmodium infection when compared to controls fed on naïve mouse blood. When we examine the expression of well-known enzymes responsible for uric acid metabolism, we see a significant increase in both urate oxidase (UO) and allatoicase (ALLC) expression following Plasmodium infection. Targeting the essential first step in uric acid metabolism by silencing UO resulted in elevated levels of uric acid, enhancing malaria parasite survival. With implications from other insect systems that bacteria can modulate UO expression, we examined the possibility that the mosquito microbiota and its expansion following blood-feeding may contribute to increased UO levels. However, there was no difference in uric acid metabolism between septic and aseptic mosquitoes, indicating that the mosquito microbiome is not associated with the manipulation of UO expression. Together, our study provides new evidence that Plasmodium infection causes the mosquito host to actively metabolize uric acid by increasing UO expression to limit Plasmodium oocyst survival, suggesting that nitrogen metabolism is an essential pathway in defining mosquito vector competence.
Collapse
|
20
|
Najjar H, Al-Ashmar S, Qush A, Al-Asmar J, Rashwan S, Elgamal A, Zeidan A, Kamareddine L. Enteric Pathogens Modulate Metabolic Homeostasis in the Drosophila melanogaster host. Microbes Infect 2022; 24:104946. [DOI: 10.1016/j.micinf.2022.104946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
|
21
|
Disparate regulation of IMD signaling drives sex differences in infection pathology in Drosophila melanogaster. Proc Natl Acad Sci U S A 2021; 118:2026554118. [PMID: 34341118 PMCID: PMC8364183 DOI: 10.1073/pnas.2026554118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sex differences in infection outcome are a widely observed phenomenon. While it is known that biological sex can influence an animal’s response to infection, the mechanisms through which these differences emerge are less clear. Here, we describe a mechanism through which heightened regulation of the IMD signaling pathway by female—but not male—Drosophila melanogaster reduces the cost of immune activity at the expense of resistance to bacterial infection. Through the masculinization of the main organ responsible for antimicrobial peptide activity in the fly (fat body), this work demonstrates that this heightened immune regulation is mediated by sex-determining pathways. Male and female animals exhibit differences in infection outcomes. One possible source of sexually dimorphic immunity is the sex-specific costs of immune activity or pathology, but little is known about the independent effects of immune- versus microbe-induced pathology and whether these may differ for the sexes. Here, by measuring metabolic and physiological outputs in Drosophila melanogaster with wild-type and mutant immune responses, we test whether the sexes are differentially impacted by these various sources of pathology and identify a critical regulator of this difference. We find that the sexes exhibit differential immune activity but similar bacteria-derived metabolic pathology. We show that female-specific immune-inducible expression of PGRP-LB, a negative regulator of the immune deficiency (IMD) pathway, enables females to reduce immune activity in response to reductions in bacterial numbers. In the absence of PGRP-LB, females are more resistant to infection, confirming the functional importance of this regulation and suggesting that female-biased immune restriction comes at a cost.
Collapse
|
22
|
Demiroz D, Platanitis E, Bryant M, Fischer P, Prchal-Murphy M, Lercher A, Lassnig C, Baccarini M, Müller M, Bergthaler A, Sexl V, Dolezal M, Decker T. Listeria monocytogenes infection rewires host metabolism with regulatory input from type I interferons. PLoS Pathog 2021; 17:e1009697. [PMID: 34237114 PMCID: PMC8266069 DOI: 10.1371/journal.ppat.1009697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/07/2021] [Indexed: 12/22/2022] Open
Abstract
Listeria monocytogenes (L. monocytogenes) is a food-borne bacterial pathogen. Innate immunity to L. monocytogenes is profoundly affected by type I interferons (IFN-I). Here we investigated host metabolism in L. monocytogenes-infected mice and its potential control by IFN-I. Accordingly, we used animals lacking either the IFN-I receptor (IFNAR) or IRF9, a subunit of ISGF3, the master regulator of IFN-I-induced genes. Transcriptomes and metabolite profiles showed that L. monocytogenes infection induces metabolic rewiring of the liver. This affects various metabolic pathways including fatty acid (FA) metabolism and oxidative phosphorylation and is partially dependent on IFN-I signaling. Livers and macrophages from Ifnar1-/- mice employ increased glutaminolysis in an IRF9-independent manner, possibly to readjust TCA metabolite levels due to reduced FA oxidation. Moreover, FA oxidation inhibition provides protection from L. monocytogenes infection, explaining part of the protection of Irf9-/- and Ifnar1-/- mice. Our findings define a role of IFN-I in metabolic regulation during L. monocytogenes infection. Metabolic differences between Irf9-/- and Ifnar1-/- mice may underlie the different susceptibility of these mice against lethal infection with L. monocytogenes.
Collapse
Affiliation(s)
- Duygu Demiroz
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
- Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and the Medical University of Vienna, Vienna, Austria
| | - Ekaterini Platanitis
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Michael Bryant
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Philipp Fischer
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Michaela Prchal-Murphy
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Alexander Lercher
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York City, New York, United States of America
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Veronika Sexl
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Decker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
23
|
Schlamp F, Delbare SYN, Early AM, Wells MT, Basu S, Clark AG. Dense time-course gene expression profiling of the Drosophila melanogaster innate immune response. BMC Genomics 2021; 22:304. [PMID: 33902461 PMCID: PMC8074482 DOI: 10.1186/s12864-021-07593-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immune responses need to be initiated rapidly, and maintained as needed, to prevent establishment and growth of infections. At the same time, resources need to be balanced with other physiological processes. On the level of transcription, studies have shown that this balancing act is reflected in tight control of the initiation kinetics and shutdown dynamics of specific immune genes. RESULTS To investigate genome-wide expression dynamics and trade-offs after infection at a high temporal resolution, we performed an RNA-seq time course on D. melanogaster with 20 time points post Imd stimulation. A combination of methods, including spline fitting, cluster analysis, and Granger causality inference, allowed detailed dissection of expression profiles, lead-lag interactions, and functional annotation of genes through guilt-by-association. We identified Imd-responsive genes and co-expressed, less well characterized genes, with an immediate-early response and sustained up-regulation up to 5 days after stimulation. In contrast, stress response and Toll-responsive genes, among which were Bomanins, demonstrated early and transient responses. We further observed a strong trade-off with metabolic genes, which strikingly recovered to pre-infection levels before the immune response was fully resolved. CONCLUSIONS This high-dimensional dataset enabled the comprehensive study of immune response dynamics through the parallel application of multiple temporal data analysis methods. The well annotated data set should also serve as a useful resource for further investigation of the D. melanogaster innate immune response, and for the development of methods for analysis of a post-stress transcriptional response time-series at whole-genome scale.
Collapse
Affiliation(s)
- Florencia Schlamp
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| | | | - Angela M Early
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Martin T Wells
- Statistics and Data Science, Cornell University, Ithaca, NY, USA
| | - Sumanta Basu
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| | - Andrew G Clark
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
24
|
Wang L, Liu C, Geng X. Identify immune-related genes of adzuki bean weevil (Callosobruchus chinensis) in response to bacteria challenge by transcriptome analysis. Microb Pathog 2021; 151:104749. [PMID: 33484809 DOI: 10.1016/j.micpath.2021.104749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Callosobruchus chinensis is one of the important postharvest pests in legume growing areas. Bacterial pesticide is a potential alternative method to control storage pests. However, the effect of these pathogen bacteria on storage pests, and the molecular mechanisms of insect response remain to be to investigated. RESULTS Using the next generation sequencing technology, we established a transcriptomic library for C. chinensis larvae in response to Escherichia coli. Total of 355 differential expressed genes (DEGs) were identified, which 178 DEGs were upregulated, and 177 DEGs were downregulated compared to control group. To validate the RNA-seq analysis, 20 DEGs and 14 immune-related genes were selected to perform quantitative polymerase chain reaction (RT-qPCR). These immune-related genes were involved in recognition (peptidoglycan recognition proteins), signal transduction (fibrinogen-related proteins, serine proteinases and NF-κB), and execution effectors (phenoloxidase, defensin, attacin, and antimicrobial peptide). In addition, genes that encode digestive and respiratory enzymes were altered in C. chinensis larvae in response to infection. Some genes that involved in juvenile hormone and insulin pathway appeared to express differentially, suggesting that pathogen infection might lead to developmental arrest. Furthermore, iron homeostasis and chitin metabolism appeared significantly altered after infection. CONCLUSION In this study, we characterized the immune response of C. chinensis larvae in response to E. coli using RNA-seq, from pathogen recognition, signal transduction, to execution. Some other identified genes were involved in iron homeostasis, respiration, and digestion. A better understanding of molecular response of beetle to pathogen will facilitate us to develop an available strategy to control storage pests.
Collapse
Affiliation(s)
- Lei Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, 212100, PR China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Chang Liu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, 212100, PR China
| | - Xueqing Geng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| |
Collapse
|
25
|
Yin H, Zhang S, Shen M, Zhang Z, Huang H, Zhao Z, Guo X, Wu P. Integrative analysis of circRNA/miRNA/mRNA regulatory network reveals the potential immune function of circRNAs in the Bombyx mori fat body. J Invertebr Pathol 2021; 179:107537. [PMID: 33472087 DOI: 10.1016/j.jip.2021.107537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Bombyx mori nucleopolyhedrosis virus (BmNPV) is one of the greatest threats to sustainable development of the sericulture industry. Circular RNA (circRNA), a type of non-coding RNA, has been shown to play important roles in gene expression regulation, immune response, and diseases. The fat body is a tissue with both metabolic and immune functions. To explore the potential immune function of circRNAs, we analyzed differentially expressed (DE)circRNAs, microRNAs(miRNAs), and mRNAs in the B. mori fat body in response to BmNPV infection using high-throughput RNA sequencing. A total of 77 DEcircRNAs, 32 DEmiRNAs, and 730 DEmRNAs that are associated with BmNPV infection were identified. We constructed a DEcircRNA/DEmiRNA/DEmRNA and DEcircRNA/DEmiRNA/BmNPV gene regulatory network and validated the differential expression of circ_0001432 and its corresponding miRNA (miR-2774c and miR-3406-5p) and mRNA (778467 and 101745232) in the network. Tissue-specific expression of circ_0001432 and its expression at different time points were also examined. KEGG pathway analysis of DEmRNAs, target genes of DEmiRNAs, and host genes of DEcircRNAs in the network showed that these genes were enriched in several metabolic pathways and signaling pathways, which could play important roles in insect immune responses. Our results suggest that circRNA could be involved in immune responses of the B. mori fat body and help in understanding the molecular mechanisms underlying silkworm-pathogen interactions.
Collapse
Affiliation(s)
- Haotong Yin
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Shaolun Zhang
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212018, China
| | - Manman Shen
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212018, China
| | - Zhengdong Zhang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212018, China
| | - Haoling Huang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Zhimeng Zhao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Xijie Guo
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212018, China
| | - Ping Wu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212018, China.
| |
Collapse
|
26
|
Sellin J, Fülle JB, Thiele C, Bauer R, Bülow MH. Free fatty acid determination as a tool for modeling metabolic diseases in Drosophila. JOURNAL OF INSECT PHYSIOLOGY 2020; 126:104090. [PMID: 32730782 DOI: 10.1016/j.jinsphys.2020.104090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 06/10/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
Free or non-esterified fatty acids are the product of lipolysis of storage fat, i.e. triacylglyceroles. When the amount of fat exceeds the capacity of lipid-storing organs, free fatty acids affect and damage other non-lipid-storing organs. This process is termed lipotoxicity. Within a cell, free fatty acids can damage mitochondria, and lipotoxicity-induced mitochondrial damage has been associated recently with Peroxisomal Biogenesis Disorders. Drosophila melanogaster has a rising popularity as a model organism for metabolic diseases, but an optimized assay for measuring free fatty acids in Drosophila tissue samples is missing. Here we present a detailed protocol highlighting technical requirements and pitfalls to determine free fatty acids in samples of Drosophila tissue. The colorimetric assay allows the reproducible and cost-efficient measurement of free fatty acids in a 96 well plate format. We used our assay to determine changes in free fatty acid levels in different developmental stages and feeding conditions, and found that larvae and adults have different patterns of free fatty acid formation during starvation. Our assay is a valuable tool in the modeling of metabolic diseases with Drosophila melanogaster.
Collapse
Affiliation(s)
- Julia Sellin
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| | - Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK; Skin Research Institute of Singapore, A*STAR, 8A Biomedical Grove, Immunos #06-06, Singapore, Singapore
| | - Christoph Thiele
- University of Bonn, Life & Medical Sciences Institute (LIMES), Biochemistry & Cell Biology of Lipids, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Reinhard Bauer
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Margret H Bülow
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| |
Collapse
|
27
|
Wang RJ, Chen K, Xing LS, Lin Z, Zou Z, Lu Z. Reactive oxygen species and antimicrobial peptides are sequentially produced in silkworm midgut in response to bacterial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 110:103720. [PMID: 32344046 DOI: 10.1016/j.dci.2020.103720] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 06/11/2023]
Abstract
The silkworm, Bombyx mori, is utilized as a research model in many aspects of biological studies, including genetics, development and immunology. Previous biochemical and genomic studies have elucidated the silkworm immunity in response to infections elicited by bacteria, fungi, microsporidia, and viruses. The intestine serves as the front line in the battle between insects and ingested harmful microorganisms. In this study, we performed RNA sequencing (RNA-seq) of the larval silkworm midgut after oral infection with the Gram-positive bacterium Bacillus bombysepticus and the Gram-negative bacterium Yersinia pseudotuberculosis. This enables us to get a comprehensive understanding of the midgut responses to bacterial infection. We found that B. bombysepticus induced much stronger immune responses than Y. pseudotuberculosis did. Bacterial infection resulted in more energy consumption including carbohydrates and fatty acids. The midgut immune system was characterized by the generation of reactive oxygen species and antimicrobial peptides. The former played a critical role in eliminating invading bacteria during early stage, while the latter executed during late stage. Our results provide an integrated insight into the midgut systematic responses to bacterial infection.
Collapse
Affiliation(s)
- Rui-Juan Wang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China.
| | - Kangkang Chen
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China.
| | - Long-Sheng Xing
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, China.
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
28
|
Allen PE, Martinez JJ. Modulation of Host Lipid Pathways by Pathogenic Intracellular Bacteria. Pathogens 2020; 9:pathogens9080614. [PMID: 32731350 PMCID: PMC7460438 DOI: 10.3390/pathogens9080614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/17/2020] [Accepted: 07/25/2020] [Indexed: 12/22/2022] Open
Abstract
Lipids are a broad group of molecules required for cell maintenance and homeostasis. Various intracellular pathogens have developed mechanisms of modulating and sequestering host lipid processes for a large array of functions for both bacterial and host cell survival. Among the host cell lipid functions that intracellular bacteria exploit for infection are the modulation of host plasma membrane microdomains (lipid rafts) required for efficient bacterial entry; the recruitment of specific lipids for membrane integrity of intracellular vacuoles; and the utilization of host lipid droplets for the regulation of immune responses and for energy production through fatty acid β-oxidation and oxidative phosphorylation. The majority of published studies on the utilization of these host lipid pathways during infection have focused on intracellular bacterial pathogens that reside within a vacuole during infection and, thus, have vastly different requirements for host lipid metabolites when compared to those intracellular pathogens that are released into the host cytosol upon infection. Here we summarize the mechanisms by which intracellular bacteria sequester host lipid species and compare the modulation of host lipid pathways and metabolites during host cell infection by intracellular pathogens residing in either a vacuole or within the cytosol of infected mammalian cells. This review will also highlight common and unique host pathways necessary for intracellular bacterial growth that could potentially be targeted for therapeutic intervention.
Collapse
|
29
|
Vincent CM, Simoes da Silva CJ, Wadhawan A, Dionne MS. Origins of Metabolic Pathology in Francisella-Infected Drosophila. Front Immunol 2020; 11:1419. [PMID: 32733472 PMCID: PMC7360822 DOI: 10.3389/fimmu.2020.01419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/02/2020] [Indexed: 12/22/2022] Open
Abstract
The origins and causes of infection pathologies are often not understood. Despite this, the study of infection and immunity relies heavily on the ability to discern between potential sources of pathology. Work in the fruit fly has supported the assumption that mortality resulting from bacterial invasion is largely due to direct host-pathogen interactions, as lower pathogen loads are often associated with reduced pathology, and bacterial load upon death is predictable. However, the mechanisms through which these interactions bring about host death are complex. Here we show that infection with the bacterium Francisella novicida leads to metabolic dysregulation and, using treatment with a bacteriostatic antibiotic, we show that this pathology is the result of direct interaction between host and pathogen. We show that mutants of the immune deficiency immune pathway fail to exhibit similar metabolic dysregulation, supporting the idea that the reallocation of resources for immune-related activities contributes to metabolic dysregulation. Targeted investigation into the cross-talk between immune and metabolic pathways has the potential to illuminate some of this interaction.
Collapse
Affiliation(s)
- Crystal M Vincent
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Carolina J Simoes da Silva
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Ashima Wadhawan
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Potts R, King JG, Pietri JE. Ex vivo characterization of the circulating hemocytes of bed bugs and their responses to bacterial exposure. J Invertebr Pathol 2020; 174:107422. [PMID: 32526226 PMCID: PMC9254597 DOI: 10.1016/j.jip.2020.107422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 11/17/2022]
Abstract
Bed bugs (Cimex spp.) are urban pests of global importance. Knowledge of the immune system of bed bugs has implications for understanding their susceptibility to biological control agents, their potential to transmit human pathogens, and the basic comparative immunology of insects. Nonetheless, the immunological repertoire of the family Cimicidae remains poorly characterized. Here, we use microscopy, flow cytometry, and RNA sequencing to provide a basal characterization of the circulating hemocytes of the common bed bug, Cimex lectularius. We also examine the responses of these specialized cells to E. coli exposure using the same techniques. Our results show that circulating hemocytes are comprised of at least four morphologically distinct cell types that are capable of phagocytosis, undergo degranulation, and exhibit additional markers of activation following stimulation, including size shift and DNA replication. Furthermore, transcriptomic profiling reveals expression of predicted Toll/IMD signaling pathway components, antimicrobial effectors and other potentially immunoresponsive genes in these cells. Together, our data demonstrate the conservation of several canonical cellular immune responses in the common bed bug and provide a foundation for additional mechanistic immunological studies with specific pathogens of interest.
Collapse
Affiliation(s)
- Rashaun Potts
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, United States
| | - Jonas G King
- Mississippi State University, Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Starkville, MS, United States
| | - Jose E Pietri
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, United States.
| |
Collapse
|
31
|
Toprak U. The Role of Peptide Hormones in Insect Lipid Metabolism. Front Physiol 2020; 11:434. [PMID: 32457651 PMCID: PMC7221030 DOI: 10.3389/fphys.2020.00434] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Lipids are the primary storage molecules and an essential source of energy in insects during reproduction, prolonged periods of flight, starvation, and diapause. The coordination center for insect lipid metabolism is the fat body, which is analogous to the vertebrate adipose tissue and liver. The fat body is primarily composed of adipocytes, which accumulate triacylglycerols in intracellular lipid droplets. Genomics and proteomics, together with functional analyses, such as RNA interference and CRISPR/Cas9-targeted genome editing, identified various genes involved in lipid metabolism and elucidated their functions. However, the endocrine control of insect lipid metabolism, in particular the roles of peptide hormones in lipogenesis and lipolysis are relatively less-known topics. In the current review, the neuropeptides that directly or indirectly affect insect lipid metabolism are introduced. The primary lipolytic and lipogenic peptide hormones are adipokinetic hormone and the brain insulin-like peptides (ILP2, ILP3, ILP5). Other neuropeptides, such as insulin-growth factor ILP6, neuropeptide F, allatostatin-A, corazonin, leucokinin, tachykinins and limostatin, might stimulate lipolysis, while diapause hormone-pheromone biosynthesis activating neuropeptide, short neuropeptide F, CCHamide-2, and the cytokines Unpaired 1 and Unpaired 2 might induce lipogenesis. Most of these peptides interact with one another, but mostly with insulin signaling, and therefore affect lipid metabolism indirectly. Peptide hormones are also involved in lipid metabolism during reproduction, flight, diapause, starvation, infections and immunity; these are also highlighted. The review concludes with a discussion of the potential of lipid metabolism-related peptide hormones in pest management.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Lab., Department of Plant Protection Ankara, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
32
|
Somerville AGT, Gleave K, Jones CM, Reimer LJ. The consequences of Brugia malayi infection on the flight and energy resources of Aedes aegypti mosquitoes. Sci Rep 2019; 9:18449. [PMID: 31804546 PMCID: PMC6895159 DOI: 10.1038/s41598-019-54819-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022] Open
Abstract
Evidence from experimental infection studies has shown that infected mosquitoes exhibit altered host-seeking behaviours, with suppression and activation of behaviours dependent on the parasite's development stage. The mechanisms are poorly characterised; however, infections can impact mosquito energy reserves, thereby influencing key life-history traits and behaviours. In addition, filarial infection is likely detrimental to flight due to damage caused by developing worms. This study aimed to evaluate the impacts of Brugia malayi infection on Aedes aegypti flight parameters: distance, average speed, maximum speed and number of flight bursts, using a tethered flight mill. In addition, we explored whether differences in flight capacity may be due to the effect of infection on glycogen and lipid reserves. Infection with filarial worms significantly reduced flight distance but increased the number of flight bursts. Exposure to microfilaermic blood led to a significant decrease in average and maximum flight speeds even in the absence of an established infection. Mosquitoes fed on microfilaraemic blood showed reduced levels of glycogen (-37.9%) and lipids (-49.7%) compared to controls at nine days post-exposure. However, a one-hour period of flight activity caused an increase in lipid content for both infected and control mosquitoes. Consequential flight incapacitation may serve in explaining the heterogeneous distribution of lymphatic filariasis.
Collapse
Affiliation(s)
| | - Katherine Gleave
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Christopher M Jones
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Lisa J Reimer
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| |
Collapse
|
33
|
Chambers MC, Jacobson E, Khalil S, Lazzaro BP. Consequences of chronic bacterial infection in Drosophila melanogaster. PLoS One 2019; 14:e0224440. [PMID: 31648237 PMCID: PMC6812774 DOI: 10.1371/journal.pone.0224440] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/14/2019] [Indexed: 11/25/2022] Open
Abstract
Even when successfully surviving an infection, a host often fails to eliminate a pathogen completely and may sustain substantial pathogen burden for the remainder of its life. Using systemic bacterial infection in Drosophila melanogaster, we characterize chronic infection by three bacterial species from different genera - Providencia rettgeri, Serratia marcescens, and Enterococcus faecalis–following inoculation with a range of doses. To assess the consequences of these chronic infections, we determined the expression of antimicrobial peptide genes, survival of secondary infection, and starvation resistance after one week of infection. While higher infectious doses unsurprisingly lead to higher risk of death, they also result in higher chronic bacterial loads among the survivors for all three infections. All three chronic infections caused significantly elevated expression of antimicrobial peptide genes at one week post-infection and provided generalized protection again secondary bacterial infection. Only P. rettgeri infection significantly influenced resistance to starvation, with persistently infected flies dying more quickly under starvation conditions relative to controls. These results suggest that there is potentially a generalized mechanism of protection against secondary infection, but that other impacts on host physiology may depend on the specific pathogen. We propose that chronic infections in D. melanogaster could be a valuable tool for studying tolerance of infection, including impacts on host physiology and behavior.
Collapse
Affiliation(s)
- Moria Cairns Chambers
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Department of Biology, Bucknell University, Lewisburg, PA, United States of America
- * E-mail:
| | - Eliana Jacobson
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
| | - Sarah Khalil
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
34
|
Loreto RG, Hughes DP. The metabolic alteration and apparent preservation of the zombie ant brain. JOURNAL OF INSECT PHYSIOLOGY 2019; 118:103918. [PMID: 31400384 DOI: 10.1016/j.jinsphys.2019.103918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 05/25/2023]
Abstract
Some parasites can manipulate the behavior of their animal hosts to increase transmission. An interesting area of research is understanding how host neurobiology is manipulated by microbes to the point of displaying such aberrant behaviors. Here, we characterize the metabolic profile of the brain of an insect at the moment of the behavioral manipulation by a parasitic microbe. Our model system are ants infected with the parasitic fungus Ophiocordyceps kimflemingiae (=unilateralis), which manipulates ants to climb and bite into plant substrates, before killing the host (i.e. zombie ants). At the moment of the behavioral manipulation by the fungus, the host's brain is not invaded by the fungus which is known to extensively invade muscle tissue. We found that, despite not being invaded by the parasite, the brains of manipulated ants are notably different, showing alterations in neuromodulatory substances, signs of neurodegeneration, changes in energy use, and antioxidant compound that signal stress reactions by the host. Ergothionine, a fungal derived compound with known neuronal cytoprotection functions was found to be highly elevated in zombie ant brains suggesting the fungus, which does not invade the central nervous system, is preserving the brain.
Collapse
Affiliation(s)
- Raquel G Loreto
- Department of Entomology and Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, 16802 PA, USA; Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Institute Pasteur, Paris 75724, France
| | - David P Hughes
- Department of Entomology and Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, 16802 PA, USA; Department of Biology, Pennsylvania State University, University Park, 16802 PA, USA.
| |
Collapse
|
35
|
Sugar Alcohols of Polyol Pathway Serve as Alarmins to Mediate Local-Systemic Innate Immune Communication in Drosophila. Cell Host Microbe 2019; 26:240-251.e8. [PMID: 31350199 DOI: 10.1016/j.chom.2019.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/30/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022]
Abstract
Interorgan immunological communication is critical to connect the local-systemic innate immune response and orchestrate a homeostatic host defense. However, the factors and their roles in this process remain unclear. We find Drosophila IMD response in guts can sequentially trigger a systemic IMD reaction in the fat body. Sugar alcohols of the polyol pathway are essential for the spatiotemporal regulation of gut-fat body immunological communication (GFIC). IMD activation in guts causes elevated levels of sorbitol and galactitol in hemolymph. Aldose reductase (AR) in hemocytes, the rate-limiting enzyme of the polyol pathway, is necessary and sufficient for the increase of plasma sugar alcohols. Sorbitol relays GFIC by subsequent activation of Metalloprotease 2, which cleaves PGRP-LC to activate IMD response in fat bodies. Thus, this work unveils how GFIC relies on the intermediate activation of the polyol pathway in hemolymph and demonstrates that AR provides a critical metabolic checkpoint in the global inflammatory response.
Collapse
|
36
|
Dolezal T, Krejcova G, Bajgar A, Nedbalova P, Strasser P. Molecular regulations of metabolism during immune response in insects. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 109:31-42. [PMID: 30959109 DOI: 10.1016/j.ibmb.2019.04.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/12/2019] [Accepted: 04/01/2019] [Indexed: 06/09/2023]
Abstract
Mounting an immune response is an energy-consuming process. Activating immune functions requires the synthesis of many new molecules and the undertaking of numerous cellular tasks and it must happen rapidly. Therefore, immune cells undergo a metabolic switch, which enables the rapid production of ATP and new biomolecules. Such metabolism is very nutrient-demanding, especially of glucose and glutamine, and thus the immune response is associated with a systemic metabolic switch, redirecting nutrient flow towards immunity and away from storage and consumption by non-immune processes. The immune system during its activation becomes privileged in terms of using organismal resources and the activated immune cells usurp nutrients by producing signals which reduce the metabolism of non-immune tissues. The insect fat body plays a dual role in which it is both a metabolic organ, storing energy and providing energy to the rest of the organism, but also an organ important for humoral immunity. Therefore, the internal switch from anabolism to the production of antimicrobial peptides occurs in the fat body during infection. The mechanisms regulating metabolism during the immune response ensure adequate energy for an effective response (resistance) but they must be properly regulated because energy is not unlimited and the energy needs of the immune system thus interfere with the needs of other physiological traits. If not properly regulated, the immune response may in the end decrease fitness via decreasing disease tolerance.
Collapse
Affiliation(s)
- Tomas Dolezal
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Gabriela Krejcova
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Adam Bajgar
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Pavla Nedbalova
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Paul Strasser
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| |
Collapse
|
37
|
Galenza A, Foley E. Immunometabolism: Insights from the Drosophila model. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 94:22-34. [PMID: 30684503 DOI: 10.1016/j.dci.2019.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
Multicellular organisms inhabit an environment that includes a mix of essential nutrients and large numbers of potentially harmful microbes. Germline-encoded receptors scan the environment for microbe associated molecular patterns, and, upon engagement, activate powerful defenses to protect the host from infection. At the same time, digestive enzymes and transporter molecules sieve through ingested material for building blocks and energy sources necessary for survival, growth, and reproduction. We tend to view immune responses as a potent array of destructive forces that overwhelm potentially harmful agents. In contrast, we view metabolic processes as essential, constructive elements in the maintenance and propagation of life. However, there is considerable evidence of functional overlap between the two processes, and disruptions to one frequently modify outputs of the other. Studies of immunometabolism, or interactions between immunity and metabolism, have increased in prominence with the discovery of inflammatory components to metabolic diseases such as type two diabetes. In this review, we will focus on contributions of studies with the fruit fly, Drosophila melanogaster, to our understanding of immunometabolism. Drosophila is widely used to study immune signaling, and to understand the regulation of metabolism in vivo, and this insect has considerable potential as a tool to build our understanding of the molecular and cellular bridges that connect immune and metabolic pathways.
Collapse
Affiliation(s)
- Anthony Galenza
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada.
| |
Collapse
|
38
|
Davoodi S, Galenza A, Panteluk A, Deshpande R, Ferguson M, Grewal S, Foley E. The Immune Deficiency Pathway Regulates Metabolic Homeostasis in Drosophila. THE JOURNAL OF IMMUNOLOGY 2019; 202:2747-2759. [DOI: 10.4049/jimmunol.1801632] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/04/2019] [Indexed: 12/28/2022]
|
39
|
The histone methyltransferase G9a regulates tolerance to oxidative stress-induced energy consumption. PLoS Biol 2019; 17:e2006146. [PMID: 30860988 PMCID: PMC6413895 DOI: 10.1371/journal.pbio.2006146] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 02/06/2019] [Indexed: 12/24/2022] Open
Abstract
Stress responses are crucial processes that require activation of genetic programs that protect from the stressor. Stress responses are also energy consuming and can thus be deleterious to the organism. The mechanisms coordinating energy consumption during stress response in multicellular organisms are not well understood. Here, we show that loss of the epigenetic regulator G9a in Drosophila causes a shift in the transcriptional and metabolic responses to oxidative stress (OS) that leads to decreased survival time upon feeding the xenobiotic paraquat. During OS exposure, G9a mutants show overactivation of stress response genes, rapid depletion of glycogen, and inability to access lipid energy stores. The OS survival deficiency of G9a mutants can be rescued by a high-sugar diet. Control flies also show improved OS survival when fed a high-sugar diet, suggesting that energy availability is generally a limiting factor for OS tolerance. Directly limiting access to glycogen stores by knocking down glycogen phosphorylase recapitulates the OS-induced survival defects of G9a mutants. We propose that G9a mutants are sensitive to stress because they experience a net reduction in available energy due to (1) rapid glycogen use, (2) an inability to access lipid energy stores, and (3) an overinduced transcriptional response to stress that further exacerbates energy demands. This suggests that G9a acts as a critical regulatory hub between the transcriptional and metabolic responses to OS. Our findings, together with recent studies that established a role for G9a in hypoxia resistance in cancer cell lines, suggest that G9a is of wide importance in controlling the cellular and organismal response to multiple types of stress. Stress responses require proper activation of genetic programs to protect the organism from the stressor. However, the mechanisms controlling energy consumption during stress responses are not well understood. Here, we investigate the role of epigenetic modifier G9a in regulating metabolism and gene transcription during oxidative stress responses in Drosophila. Flies lacking G9a show a shift in the metabolic and transcriptional responses to oxidative stress, leading to decreased stress tolerance despite intact oxidative stress defense mechanisms. During oxidative stress exposure, G9a mutants show overactivation of stress response and many other genes, rapid depletion of glycogen energy stores, and an inability to access lipid energy stores. The increased susceptibility of G9a mutant flies to oxidative stress can be rescued simply by providing extra sugar. This suggests that G9a mutants are sensitive to stress because of reduced access to immediately available energy. Wild-type flies also become more tolerant to oxidative stress when they are fed extra sugar, whereas blocking energy access by genetically reducing a key metabolic enzyme leads to oxidative stress sensitivity. Though the genetic response to oxidative stress has long been appreciated, our study emphasizes the importance of energy metabolism for stress tolerance and identifies the histone methyltransferase G9a as an important player regulating both.
Collapse
|
40
|
Yang KH, Yun B, Choi HJ, Ryu S, Lee WJ, Oh MH, Song MH, Kim JN, Oh S, Kim Y, Kim YJ. Simple Evaluation of Listeria monocytogenes Pathogenesis Using Caenorhabditis elegans Animal Model. Food Sci Anim Resour 2019; 39:84-92. [PMID: 30882077 PMCID: PMC6411236 DOI: 10.5851/kosfa.2019.e6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/31/2018] [Accepted: 01/02/2019] [Indexed: 11/06/2022] Open
Abstract
Listeria monocytogenes is a major cause of serious foodborne illness in the dairy foods. Although Caenorhabditis elegans model is well established as a virulence model of pathogenic bacteria, its application on L. monocytogenes is critically unclear. The objective of this study was to carry out an evaluation of L. monocytogenes toxicity using C. elegans nematode as a simple host model. We found that C. elegans nematodes have high susceptibility to L. monocytogenes infection, as a consequence of accumulation of bacteria in the worms' intestine. However, L. innocua, which is known to be non-toxic, is not accumulate in the intestine of worms and is not toxic similarly to Escherichia coli OP50 known as the normal feed source of C. elegans. Importantly, immune-associated genes of C. elegans were intensely upregulated more than 3.0-fold when they exposed to L. monocytogenes. In conclusion, we established that C. elegans is an effective model for studying the toxicity of L. monocytogenes and we anticipate that this system will result in the discovery of many potential anti-listeria agents for dairy foods.
Collapse
Affiliation(s)
- Kyoung Hee Yang
- Department of Food and Biotechnology,
Korea University, Sejong 30019,
Korea
| | - Bohyun Yun
- Department of Animal Science and Institute
of Milk Genomics, Chonbuk National University,
Jeonju 54896, Korea
| | - Hye Jin Choi
- Department of Animal Science and Institute
of Milk Genomics, Chonbuk National University,
Jeonju 54896, Korea
| | - Sangdon Ryu
- Department of Animal Science and Institute
of Milk Genomics, Chonbuk National University,
Jeonju 54896, Korea
| | - Woong Ji Lee
- Department of Animal Science and Institute
of Milk Genomics, Chonbuk National University,
Jeonju 54896, Korea
| | - Mi-Hwa Oh
- Animal Products Research and Development
Division, National Institute of Animal Science, Rural Development
Administration, Wanju 55365,
Korea
| | - Min-Ho Song
- Division of Animal and Dairy Science,
Chungnam National University, Daejeon 34134,
Korea
| | - Jong Nam Kim
- Department of Beef Science, Korean
National College of Agriculture and Fisheries,
Jeonju 54874, Korea
| | - Sangnam Oh
- Department of Functional Food and
Biotechnology, Jeonju University, Jeonju 55069,
Korea
| | - Younghoon Kim
- Department of Animal Science and Institute
of Milk Genomics, Chonbuk National University,
Jeonju 54896, Korea
| | - Young Jun Kim
- Department of Food and Biotechnology,
Korea University, Sejong 30019,
Korea
| |
Collapse
|
41
|
Shaw DK, Tate AT, Schneider DS, Levashina EA, Kagan JC, Pal U, Fikrig E, Pedra JHF. Vector Immunity and Evolutionary Ecology: The Harmonious Dissonance. Trends Immunol 2018; 39:862-873. [PMID: 30301592 PMCID: PMC6218297 DOI: 10.1016/j.it.2018.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022]
Abstract
Recent scientific breakthroughs have significantly expanded our understanding of arthropod vector immunity. Insights in the laboratory have demonstrated how the immune system provides resistance to infection, and in what manner innate defenses protect against a microbial assault. Less understood, however, is the effect of biotic and abiotic factors on microbial-vector interactions and the impact of the immune system on arthropod populations in nature. Furthermore, the influence of genetic plasticity on the immune response against vector-borne pathogens remains mostly elusive. Herein, we discuss evolutionary forces that shape arthropod vector immunity. We focus on resistance, pathogenicity and tolerance to infection. We posit that novel scientific paradigms should emerge when molecular immunologists and evolutionary ecologists work together.
Collapse
Affiliation(s)
- Dana K Shaw
- Department of Veterinary Microbiology and Pathology, Washington State, Pullman, WA, USA.
| | - Ann T Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| | - David S Schneider
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Elena A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
42
|
Harsh S, Ozakman Y, Kitchen SM, Paquin-Proulx D, Nixon DF, Eleftherianos I. Dicer-2 Regulates Resistance and Maintains Homeostasis against Zika Virus Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2018; 201:3058-3072. [PMID: 30305326 DOI: 10.4049/jimmunol.1800597] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) outbreaks pose a massive public health threat in several countries. We have developed an in vivo model to investigate the host-ZIKV interaction in Drosophila We have found that a strain of ZIKV replicates in wild-type flies without reducing their survival ability. We have shown that ZIKV infection triggers RNA interference and that mutating Dicer-2 results in enhanced ZIKV load and increased susceptibility to ZIKV infection. Using a flavivirus-specific Ab, we have found that ZIKV is localized in the gut and fat body cells of the infected wild-type flies and results in their perturbed homeostasis. In addition, Dicer-2 mutants display severely reduced insulin activity, which could contribute toward the increased mortality of these flies. Our work establishes the suitability of Drosophila as the model system to study host-ZIKV dynamics, which is expected to greatly advance our understanding of the molecular and physiological processes that determine the outcome of this disease.
Collapse
Affiliation(s)
- Sneh Harsh
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Yaprak Ozakman
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Shannon M Kitchen
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Dominic Paquin-Proulx
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Douglas F Nixon
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| |
Collapse
|
43
|
Lee KA, Lee WJ. Immune-metabolic interactions during systemic and enteric infection in Drosophila. CURRENT OPINION IN INSECT SCIENCE 2018; 29:21-26. [PMID: 30551821 DOI: 10.1016/j.cois.2018.05.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/23/2018] [Indexed: 06/09/2023]
Abstract
Immune-metabolic interactions are evolutionarily conserved phenomena observed in all metazoans, from invertebrates to vertebrates. Although it is believed that immune activation drives chronic metabolic diseases, clear conclusions about the reasons and mechanisms of cross-talk between immune and metabolic signaling cannot be drawn. The Drosophila insect model equipped with genetically well-defined immune and metabolic signaling pathways is suitable in understanding the molecular codes underlying immune-metabolic interactions. In this report, we present and discuss the following: how immune signaling induces metabolic phenotypes and how metabolic signaling sequentially induces immune phenotypes in Drosophila. Further genetic studies on immune-metabolic interactions in Drosophila are warranted to increase the understanding of the etiology of different dysregulated immune-metabolic interaction-caused diseases.
Collapse
Affiliation(s)
- Kyung-Ah Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; National Creative Research Initiative Center for Hologenomics, Seoul National University, Seoul 08826, South Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea
| | - Won-Jae Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; National Creative Research Initiative Center for Hologenomics, Seoul National University, Seoul 08826, South Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
44
|
Lissner MM, Schneider DS. The physiological basis of disease tolerance in insects. CURRENT OPINION IN INSECT SCIENCE 2018; 29:133-136. [PMID: 30551820 DOI: 10.1016/j.cois.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 06/09/2023]
Abstract
Immunology textbooks teach us about the ways hosts can recognize and kill microbes but leave out something important: the mechanisms used to survive infections. Survival depends on more than simply detecting and eliminating microbes; it requires that we prevent and repair the damage caused by pathogens and the immune response. Recent work in insects is helping to build our understanding of this aspect of pathology, called disease tolerance. Here we discuss papers that explore disease tolerance using theoretical, population genetics, and mechanistic approaches.
Collapse
Affiliation(s)
- Michelle M Lissner
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, United States
| | - David S Schneider
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
45
|
Reddy VP, Chinta KC, Saini V, Glasgow JN, Hull TD, Traylor A, Rey-Stolle F, Soares MP, Madansein R, Rahman MA, Barbas C, Nargan K, Naidoo T, Ramdial PK, George JF, Agarwal A, Steyn AJC. Ferritin H Deficiency in Myeloid Compartments Dysregulates Host Energy Metabolism and Increases Susceptibility to Mycobacterium tuberculosis Infection. Front Immunol 2018; 9:860. [PMID: 29774023 PMCID: PMC5943674 DOI: 10.3389/fimmu.2018.00860] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/06/2018] [Indexed: 12/20/2022] Open
Abstract
Iron is an essential factor for the growth and virulence of Mycobacterium tuberculosis (Mtb). However, little is known about the mechanisms by which the host controls iron availability during infection. Since ferritin heavy chain (FtH) is a major intracellular source of reserve iron in the host, we hypothesized that the lack of FtH would cause dysregulated iron homeostasis to exacerbate TB disease. Therefore, we used knockout mice lacking FtH in myeloid-derived cell populations to study Mtb disease progression. We found that FtH plays a critical role in protecting mice against Mtb, as evidenced by increased organ burden, extrapulmonary dissemination, and decreased survival in Fth-/- mice. Flow cytometry analysis showed that reduced levels of FtH contribute to an excessive inflammatory response to exacerbate disease. Extracellular flux analysis showed that FtH is essential for maintaining bioenergetic homeostasis through oxidative phosphorylation. In support of these findings, RNAseq and mass spectrometry analyses demonstrated an essential role for FtH in mitochondrial function and maintenance of central intermediary metabolism in vivo. Further, we show that FtH deficiency leads to iron dysregulation through the hepcidin-ferroportin axis during infection. To assess the clinical significance of our animal studies, we performed a clinicopathological analysis of iron distribution within human TB lung tissue and showed that Mtb severely disrupts iron homeostasis in distinct microanatomic locations of the human lung. We identified hemorrhage as a major source of metabolically inert iron deposition. Importantly, we observed increased iron levels in human TB lung tissue compared to healthy tissue. Overall, these findings advance our understanding of the link between iron-dependent energy metabolism and immunity and provide new insight into iron distribution within the spectrum of human pulmonary TB. These metabolic mechanisms could serve as the foundation for novel host-directed strategies.
Collapse
Affiliation(s)
- Vineel P. Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Krishna C. Chinta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vikram Saini
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joel N. Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Travis D. Hull
- Division of Cardiothoracic Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amie Traylor
- Nephrology Research and Training Center, University of Alabama at Birmingham and Birmingham VA Medical Center, Birmingham, AL, United States
| | - Fernanda Rey-Stolle
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | | | - Rajhmun Madansein
- Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | | | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Kievershen Nargan
- Department of Anatomical Pathology, National Health Laboratory Service, University of KwaZulu-Natal, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - Threnesan Naidoo
- Department of Anatomical Pathology, National Health Laboratory Service, University of KwaZulu-Natal, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - Pratistadevi K. Ramdial
- Department of Anatomical Pathology, National Health Laboratory Service, University of KwaZulu-Natal, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - James F. George
- Division of Cardiothoracic Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anupam Agarwal
- Nephrology Research and Training Center, University of Alabama at Birmingham and Birmingham VA Medical Center, Birmingham, AL, United States
| | - Adrie J. C. Steyn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Africa Health Research Institute (AHRI), Durban, South Africa
- UAB Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
46
|
Thioester-Containing Proteins 2 and 4 Affect the Metabolic Activity and Inflammation Response in Drosophila. Infect Immun 2018; 86:IAI.00810-17. [PMID: 29463615 DOI: 10.1128/iai.00810-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/06/2018] [Indexed: 12/17/2022] Open
Abstract
Drosophila melanogaster is an outstanding model for studying host antipathogen defense. Although substantial progress has been made in understanding how metabolism and immunity are interrelated in flies, little information has been obtained on the molecular players that regulate metabolism and inflammation in Drosophila during pathogenic infection. Recently, we reported that the inactivation of thioester-containing protein 2 (Tep2) and Tep4 promotes survival and decreases the bacterial burden in flies upon infection with the virulent pathogens Photorhabdus luminescens and Photorhabdus asymbiotica Here, we investigated physiological and pathological defects in tep mutant flies in response to Photorhabdus challenge. We find that tep2 and tep4 loss-of-function mutant flies contain increased levels of carbohydrates and triglycerides in the presence or absence of Photorhabdus infection. We also report that Photorhabdus infection leads to higher levels of nitric oxide and reduced transcript levels of the apical caspase-encoding gene Dronc in tep2 and tep4 mutants. We show that Tep2 and Tep4 are upregulated mainly in the fat body rather than the gut in Photorhabdus-infected wild-type flies and that tep mutants contain decreased numbers of Photorhabdus bacteria in both tissue types. We propose that the inactivation of Tep2 or Tep4 in adult Drosophila flies results in lower levels of inflammation and increased energy reserves in response to Photorhabdus, which could confer a survival-protective effect during the initial hours of infection.
Collapse
|
47
|
Lee KA, Cho KC, Kim B, Jang IH, Nam K, Kwon YE, Kim M, Hyeon DY, Hwang D, Seol JH, Lee WJ. Inflammation-Modulated Metabolic Reprogramming Is Required for DUOX-Dependent Gut Immunity in Drosophila. Cell Host Microbe 2018; 23:338-352.e5. [DOI: 10.1016/j.chom.2018.01.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/07/2017] [Accepted: 01/23/2018] [Indexed: 12/22/2022]
|
48
|
Abstract
Here, we provide a brief review of the mechanistic connections between immunity and aging—a fundamental biological relationship that remains poorly understood—by considering two intertwined questions: how does aging affect immunity, and how does immunity affect aging? On the one hand, aging contributes to the deterioration of immune function and predisposes the organism to infections (“immuno-senescence”). On the other hand, excessive activation of the immune system can accelerate degenerative processes, cause inflammation and immunopathology, and thus promote aging (“inflammaging”). Interestingly, several recent lines of evidence support the hypothesis that restrained or curbed immune activity at old age (that is, optimized age-dependent immune homeostasis) might actually improve realized immune function and thereby promote longevity. We focus mainly on insights from
Drosophila, a powerful genetic model system in which both immunity and aging have been extensively studied, and conclude by outlining several unresolved questions in the field.
Collapse
Affiliation(s)
- Kathrin Garschall
- Department of Ecology & Evolution, University of Lausanne, Lausanne, Switzerland
| | - Thomas Flatt
- Department of Ecology & Evolution, University of Lausanne, Lausanne, Switzerland.,Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
49
|
Metabolomics: State-of-the-Art Technologies and Applications on Drosophila melanogaster. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:257-276. [PMID: 29951824 DOI: 10.1007/978-981-13-0529-0_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metabolomics is one of the latest "omics" technology concerned with the high-throughput identification and quantification of metabolites, the final products of cellular processes. The revealed data provide an instantaneous snapshot of an organism's metabolic pathways, which can be used to explain its phenotype or physiology. On the other hand, Drosophila has shown its power in studying metabolism and related diseases. At this stage, we have the state-of-the-art knowledge in place: a potential candidate to study cellular metabolism (Drosophila melanogaster) and a powerful methodology for metabolic network decipherer (metabolomics). Yet missing is advanced metabolomics technologies like isotope-assisted metabolomics optimized for Drosophila. In this chapter, we will discuss on the current status and future perspectives in technologies and applications of Drosophila metabolomics.
Collapse
|
50
|
Bülow MH, Wingen C, Senyilmaz D, Gosejacob D, Sociale M, Bauer R, Schulze H, Sandhoff K, Teleman AA, Hoch M, Sellin J. Unbalanced lipolysis results in lipotoxicity and mitochondrial damage in peroxisome-deficient Pex19 mutants. Mol Biol Cell 2017; 29:396-407. [PMID: 29282281 PMCID: PMC6014165 DOI: 10.1091/mbc.e17-08-0535] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 01/01/2023] Open
Abstract
Peroxisomal dysfunction is often associated with mitochondrial abnormalities for unknown reasons. We found that peroxisomal loss upon Pex19 mutation in Drosophila results in Hnf4 hyperactivation with free fatty acid accumulation and mitochondrial damage as a consequence. Genetic reduction of Hnf4 in Pex19 mutants improves all phenotypes, including lethality. Inherited peroxisomal biogenesis disorders (PBDs) are characterized by the absence of functional peroxisomes. They are caused by mutations of peroxisomal biogenesis factors encoded by Pex genes, and result in childhood lethality. Owing to the many metabolic functions fulfilled by peroxisomes, PBD pathology is complex and incompletely understood. Besides accumulation of peroxisomal educts (like very-long-chain fatty acids [VLCFAs] or branched-chain fatty acids) and lack of products (like bile acids or plasmalogens), many peroxisomal defects lead to detrimental mitochondrial abnormalities for unknown reasons. We generated Pex19 Drosophila mutants, which recapitulate the hallmarks of PBDs, like absence of peroxisomes, reduced viability, neurodegeneration, mitochondrial abnormalities, and accumulation of VLCFAs. We present a model of hepatocyte nuclear factor 4 (Hnf4)-induced lipotoxicity and accumulation of free fatty acids as the cause for mitochondrial damage in consequence of peroxisome loss in Pex19 mutants. Hyperactive Hnf4 signaling leads to up-regulation of lipase 3 and enzymes for mitochondrial β-oxidation. This results in enhanced lipolysis, elevated concentrations of free fatty acids, maximal β-oxidation, and mitochondrial abnormalities. Increased acid lipase expression and accumulation of free fatty acids are also present in a Pex19-deficient patient skin fibroblast line, suggesting the conservation of key aspects of our findings.
Collapse
Affiliation(s)
- Margret H Bülow
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Christian Wingen
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Deniz Senyilmaz
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Dominic Gosejacob
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Mariangela Sociale
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Reinhard Bauer
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Heike Schulze
- Department of Membrane Biology & Lipid Biochemistry, Life & Medical Sciences Institute (LIMES), Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, 53121 Bonn, Germany
| | - Konrad Sandhoff
- Department of Membrane Biology & Lipid Biochemistry, Life & Medical Sciences Institute (LIMES), Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, 53121 Bonn, Germany
| | - Aurelio A Teleman
- Division of Signal Transduction in Cancer and Metabolism, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Michael Hoch
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Julia Sellin
- Department of Molecular Developmental Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| |
Collapse
|