1
|
Rommelaere S, Schüpfer F, Armand F, Hamelin R, Lemaitre B. An updated proteomic analysis of Drosophila haemolymph after bacterial infection. Fly (Austin) 2025; 19:2485685. [PMID: 40223358 PMCID: PMC12005426 DOI: 10.1080/19336934.2025.2485685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Using an in-depth Mass Spectrometry-based proteomics approach, we provide a comprehensive characterization of the hemolymphatic proteome of adult flies upon bacterial infection. We detected and quantified changes in abundance of several known immune regulators and effectors, including multiple antimicrobial peptides, peptidoglycan-binding proteins and serine proteases. Comparison to previously published transcriptomic analyses reveals a partial overlap with our dataset, indicating that many proteins released into the haemolymph upon infection may not be regulated at the transcript level. Among them, we identify a set of muscle-derived proteins released into the haemolymph upon infection. Finally, our analysis reveals that infection induces major changes in the abundance of proteins associated with mitochondrial respiration. This study uncovers a large number of previously undescribed proteins potentially involved in the immune response.
Collapse
Affiliation(s)
- Samuel Rommelaere
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fanny Schüpfer
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Florence Armand
- EPFL Proteomics Core Facility, EPFL SV PTECH PTP, Lausanne, Switzerland
| | - Romain Hamelin
- EPFL Proteomics Core Facility, EPFL SV PTECH PTP, Lausanne, Switzerland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
2
|
Park JS, Sung MJ, Na HJ. Drosophila model systems reveal intestinal stem cells as key players in aging. Ann N Y Acad Sci 2025; 1547:88-99. [PMID: 40276941 DOI: 10.1111/nyas.15351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The intestines play important roles in responding immediately and dynamically to food intake, environmental stress, and metabolic dysfunction, and they are involved in various human diseases and aging. A key part of their function is governed by intestinal stem cells (ISCs); therefore, understanding ISCs is vital. Dysregulation of ISC activity, which is influenced by various cell signaling pathways and environmental signals, can lead to inflammatory responses, tissue damage, and increased cancer susceptibility. Aging exacerbates these dynamics and affects ISC function and tissue elasticity. Additionally, proliferation and differentiation profoundly affect ISC behavior and gut health, highlighting the complex interplay between environmental factors and gut homeostasis. Drosophila models help us understand the complex regulatory networks in the gut, providing valuable insights into disease mechanisms and therapeutic strategies targeting human intestinal diseases.
Collapse
Affiliation(s)
- Joung-Sun Park
- Institute of Nanobio Convergence, Pusan National University, Busan, Republic of Korea
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Mi Jeong Sung
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hyun-Jin Na
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| |
Collapse
|
3
|
He L, Zhang Y, Li J, Chen D, Yue S, Liu Y, Guo Y, Wang Y, Xiu M, He J. Dunhuang Dabupi Decoction and its active components alleviate ulcerative colitis by activating glutathione metabolism and inhibiting JAK-STAT pathway in Drosophila and mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119717. [PMID: 40164365 DOI: 10.1016/j.jep.2025.119717] [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: 02/08/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dabupi Decoction (DBPD) originates from the ancient Dunhuang medical literature "Fu Xing Jue Visceral to Drug law legend" for more than 1000 years, which has been extensively employed to treat various diseases related to the spleen and stomach. However, limited studies focus on the mechanism of DBPD against ulcerative colitis (UC). AIM OF THE STUDY The beneficial effect and mechanism of DBPD against UC were detected by adopting both Drosophila melanogaster and C57BL/6J mouse models. METHODS The protective effect of DBPD against DSS-induced intestinal damage in flies was investigated by utilizing survival rate, locomotion, excretion, smurf, intestinal length, intestinal acid-base homeostasis, and Tepan blue assay. In mice, HE staining and ELISA kit were employed to assess serum histopathological damage and inflammatory factor levels. Subsequently, the molecular mechanism of DBPD was subsequently detected via DHE staining, immunofluorescence, transmission electron microscopy (TEM), real-time PCR, and transcriptomic sequencing. Additionally, liquid chromatography-mass spectrometry (LC-MS) and phenotype experiments in UC flies were utilized to identify the bioactive components of DBPD against UC. RESULTS Oral administration of DBPD remarkably alleviated DSS-induced body damage in flies by improving survival rate, locomotion, and excretion. It also remarkably rescued intestinal morphological damage, repaired acid-base homeostatic imbalance, inhibited intestinal epithelial cells (IECs) death and excessive proliferation of intestinal stem cells (ISCs), and improved ultrastructural damage of IECs in flies treated with DSS. Consistently, DBPD attenuated colitis symptoms, alleviated intestinal histopathological damage, and restored the expression of inflammatory factors in DSS-induced UC mice. As suggested by an integration of transcriptome data with molecular biology experiments, DBPD not only dramatically alleviated oxidative damage by activating the glutathione metabolic pathway, but also lowered inflammatory reaction by inhibiting the JAK-STAT pathway. Additionally, four compounds of DBPD, rhein acid, isoquercitrin, curcumin, and zeaxanthin were identified to alleviate the DSS-induced intestinal injury. CONCLUSION DBPD demonstrate immense potential for intestinal injury predominantly by activating the glutathione metabolic pathway to alleviate oxidative damage, and inhibiting the JAK-STAT pathway to mitigate inflammatory response. Rhein acid, isoquercitrin, curcumin, and zeaxanthin were the bioactive compounds of DBPD against UC.
Collapse
Affiliation(s)
- Li He
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Yongxuan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Jiangnan Li
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Dandan Chen
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Shiqi Yue
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China.
| | - Yaqiong Guo
- Second Provincial People's Hospital of Gansu, Lanzhou, 730000, China.
| | - Yan Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| | - Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China.
| | - Jianzheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, 730000, China; Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Kumar S, D'Souza LC, Shaikh FH, Rathor P, Ratnasekhar CH, Sharma A. Multigenerational immunotoxicity assessment: A three-generation study in Drosophila melanogaster upon developmental exposure to triclosan. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 370:125860. [PMID: 39954761 DOI: 10.1016/j.envpol.2025.125860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Triclosan (TCS) is widely used as an antibacterial agent, nevertheless, its presence in different environmental matrices and its persistent environmental nature pose a significant threat to the organism, including humans. Numerous studies showed that TCS exposure could lead to multiple toxicities, including immune dysfunction. However, whether parental TCS exposure could impair the offspring's immune response remains limited. Maintaining the immune homeostasis is imperative to neutralize the pathogen and crucial for tissue repair and the organism's survival. Thus, this study aimed to assess the multigenerational immune response of TCS using Drosophila melanogaster. TCS was administered to organisms (1.0, 10, and 100.0 μg/mL) over three generations during their developing phases, and its effect on the immunological response of the unexposed progeny was evaluated. Total circulatory hemocyte (immune cells) count, crystal cell count, phagocytic activity, clotting time, gene expression related to immune response and epigenetics, ROS generation, and cell death were assessed in the offspring. A concentration-dependent decline in total hemocytes, crystal cells, phagocytic activity, and increased clotting time in the subsequent generations was observed. Furthermore, parental TCS exposure enhanced the ROS levels, induced cell death, and altered the expression of antimicrobial peptides drosomycin, diptericin, and inflammatory genes upd1, upd2, and upd3, in the offspring's hemocytes across successive generations. The upregulation of reaper hid, and grim suggests that TCS promotes apoptotic death in the offspring's hemocytes. Notably, the increased mRNA expression of epigenetic regulators dnmt2 and g9a in the hemocytes of the offspring indicates epigenetic modifications. Further, we also observed that the antioxidant N-acetylcysteine (NAC) supplementation to the parents alleviated TCS toxicity and improved immunological functions in the progeny, indicating the role of ROS in the TCS-induced multigenerational immune toxicity. This finding provides valuable insights into the potential immune risk of prenatal TCS exposure to their offspring in the higher organism.
Collapse
Affiliation(s)
- Sandeep Kumar
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Faiz Hanif Shaikh
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Priya Rathor
- Metabolomics Lab, Council of Scientific and Industrial Research (CSIR)-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - C H Ratnasekhar
- Metabolomics Lab, Council of Scientific and Industrial Research (CSIR)-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
5
|
Cheung S, Bredikhin D, Gerber T, Steenbergen PJ, Basu S, Bailleul R, Hansen P, Paix A, Benton MA, Korswagen HC, Arendt D, Stegle O, Ikmi A. Systemic coordination of whole-body tissue remodeling during local regeneration in sea anemones. Dev Cell 2025; 60:780-793.e7. [PMID: 39615481 DOI: 10.1016/j.devcel.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/06/2024] [Accepted: 11/01/2024] [Indexed: 03/14/2025]
Abstract
The complexity of regeneration extends beyond local wound responses, eliciting systemic processes across the entire organism. However, the functional relevance and coordination of distant molecular processes remain unclear. In the cnidarian Nematostella vectensis, we show that local regeneration triggers a systemic homeostatic response, leading to coordinated whole-body remodeling. Leveraging spatial transcriptomics, endogenous protein tagging, and live imaging, we comprehensively dissect this systemic response at the organismal scale. We identify proteolysis as a critical process driven by both local and systemic upregulation of metalloproteases. We show that metalloproteinase expression levels and activity scale with the extent of tissue loss. This proportional response drives long-range tissue and extracellular matrix movement. Our findings demonstrate the adaptive nature of the systematic response in regeneration, enabling the organism to maintain shape homeostasis while coping with a wide range of injuries.
Collapse
Affiliation(s)
- Stephanie Cheung
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Danila Bredikhin
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Tobias Gerber
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany; European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany
| | - Petrus J Steenbergen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Soham Basu
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Richard Bailleul
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Pauline Hansen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Alexandre Paix
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Matthew A Benton
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Detlev Arendt
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Oliver Stegle
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, Cambridgeshire, UK.
| | - Aissam Ikmi
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany.
| |
Collapse
|
6
|
Cardoso-Jaime V, Dimopoulos G. Anopheles gambiae phagocytic hemocytes promote Plasmodium falciparum infection by regulating midgut epithelial integrity. Nat Commun 2025; 16:1465. [PMID: 39920122 PMCID: PMC11805967 DOI: 10.1038/s41467-025-56313-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
For successful transmission, the malaria parasite must traverse tissue epithelia and survive attack from the insect's innate immune system. Hemocytes play a multitude of roles in mosquitoes, including defense against invading pathogens. Here, we show that hemocytes of the major malaria vector Anopheles gambiae promote Plasmodium falciparum infection by maintaining midgut epithelial integrity by controlling cell proliferation upon blood feeding. The mosquito's hemocytes also control the midgut microbiota and immune gene expression. Our study unveils novel hemocyte functions that are exploited by the human malaria parasite to evade the mosquito's immune system.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
7
|
Zhang Q, Zhou X, Feng T, Tong H, Wang J, Dai J. The immune function of thioester-containing proteins in typical invertebrate disease vectors. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 176:104218. [PMID: 39579796 DOI: 10.1016/j.ibmb.2024.104218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Disease vectors, such as arthropods, primarily rely on innate immunity to counteract pathogen invasions, typically through the recognition and binding of pathogen-associated molecular patterns (PAMPs) by the host's pattern recognition receptors (PRRs). As a conserved immune effector gene family from insects to mammals, the complement system may play an essential role in combating pathogenic microorganisms. In arthropods, the complement proteins are often referred to as thioester-containing proteins (TEPs) because thioester motifs are one of the essential functional domains of the first proteins characterized within the C3 and A2M family. TEPs mainly function as specialized PRRs in sensing and binding to pathogens or their components. This paper presents a comprehensive review of the common domain and functions of TEPs in major disease vectors, in particular the specific decision-making ones expressed by Arthropoda (medical arthropods) and Mollusca (Biomphalaria glabrata) after pathogen infections. The relationship between the structure and antibacterial/antiviral activities of TEPs would further our understandings on the mechanisms governing the initiation of innate immune responses in typical disease vectors.
Collapse
Affiliation(s)
- Qianqian Zhang
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xia Zhou
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Tingting Feng
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Tong
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Jianfeng Dai
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
8
|
Touré H, Durand N, Rincheval V, Girard-Misguich F, Guénal I, Herrmann JL, Szuplewski S. Remote disruption of intestinal homeostasis by Mycobacterium abscessus is detrimental to Drosophila survival. Sci Rep 2024; 14:30775. [PMID: 39730463 DOI: 10.1038/s41598-024-80994-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/21/2024] [Indexed: 12/29/2024] Open
Abstract
Mycobacterium abscessus (Mabs), an intracellular and opportunistic pathogen, is considered the most pathogenic fast-growing mycobacterium, and causes severe pulmonary infections in patients with cystic fibrosis. While bacterial factors contributing to its pathogenicity are well studied, the host factors and responses that worsen Mabs infection are not fully understood. Here, we report that Mabs systemic infection alters Drosophila melanogaster intestinal homeostasis. Mechanistically, Mabs remotely induces a self-damaging oxidative burst, leading to excessive differentiation of intestinal stem cells into enterocytes. We demonstrated that the subsequent increased intestinal renewal is mediated by both the Notch and JAK/STAT pathways and is deleterious to Drosophila survival. In conclusion, this work highlights that the ability of Mabs to induce an exacerbated and self-damaging response in the host contributes to its pathogenesis.
Collapse
Affiliation(s)
- Hamadoun Touré
- Infection et Inflammation, Université Paris-Saclay, UVSQ, INSERM, 78180, Montigny-Le-Bretonneux, France.
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.
| | - Nicolas Durand
- Infection et Inflammation, Université Paris-Saclay, UVSQ, INSERM, 78180, Montigny-Le-Bretonneux, France
| | | | - Fabienne Girard-Misguich
- Infection et Inflammation, Université Paris-Saclay, UVSQ, INSERM, 78180, Montigny-Le-Bretonneux, France
| | - Isabelle Guénal
- Université Paris-Saclay, UVSQ, LGBC, 78000, Versailles, France
| | - Jean-Louis Herrmann
- Infection et Inflammation, Université Paris-Saclay, UVSQ, INSERM, 78180, Montigny-Le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, 92380, Garches, France
| | | |
Collapse
|
9
|
Wang X, Bao H, Huang YC, Barua A, Lai CM, Sun J, Zhou Y, Cong F, Gong S, Chang CH, Deng WM. Sex-dimorphic tumor growth is regulated by tumor microenvironmental and systemic signals. SCIENCE ADVANCES 2024; 10:eads4229. [PMID: 39642218 PMCID: PMC11623276 DOI: 10.1126/sciadv.ads4229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/01/2024] [Indexed: 12/08/2024]
Abstract
Tumor growth and progression involve coordinated regulation by internal, microenvironmental, and systemic signals and often display conspicuous sexual dimorphism. The mechanisms governing the integration and coordination of these signals, along with their sex-based differences, remain largely unknown. Using a Drosophila tumor model originating from nonreproductive tissue, we show that female-biased tumor growth involves multifaceted communications among tumor cells, hemocytes, and neuroendocrine insulin-producing cells (IPCs). Notch-active tumor cells recruit hemocytes carrying the tumor necrosis factor-α (TNF-α) homolog Eiger to the tumor microenvironment (TME), activating the c-Jun N-terminal kinase (JNK) pathway in tumor cells, instigating the sexually dimorphic up-regulation of cytokine Unpaired 2 (Upd2). Upd2, in turn, exerts a distal influence by modulating the release of a Drosophila insulin-like peptide (Dilp2) from IPCs. Dilp2 then activates the insulin signaling in the tumor, thereby fostering sexual-dimorphic tumor growth. Together, these findings reveal a relay mechanism involving the TME and systemic signals that collectively control the sexual dimorphism of tumor growth.
Collapse
Affiliation(s)
- Xianfeng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Hongcun Bao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Anindita Barua
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | - Jie Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Youfang Zhou
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Fei Cong
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | | | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Trubin S, Patel DB, Tian A. Regulation of the Intestinal Stem Cell Pool and Proliferation in Drosophila. Cells 2024; 13:1856. [PMID: 39594605 PMCID: PMC11592481 DOI: 10.3390/cells13221856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Understanding the regulation of somatic stem cells, both during homeostasis and in response to environmental challenges like injury, infection, chemical exposure, and nutritional changes, is critical because their dysregulation can result in tissue degeneration or tumorigenesis. The use of models such as the Drosophila and mammalian adult intestines offers valuable insights into tissue homeostasis and regeneration, advancing our knowledge of stem cell biology and cancer development. This review highlights significant findings from recent studies, unveiling the molecular mechanisms that govern self-renewal, proliferation, differentiation, and regeneration of intestinal stem cells (ISCs). These insights not only enhance our understanding of normal tissue maintenance but also provide critical perspectives on how ISC dysfunction can lead to pathological conditions such as colorectal cancer (CRC).
Collapse
Affiliation(s)
- Simona Trubin
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Dhruv B. Patel
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Aiguo Tian
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
- Tulane Aging Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
11
|
Vidal M, Arch M, Fuentes E, Cardona PJ. Drosophila melanogaster experimental model to test new antimicrobials: a methodological approach. Front Microbiol 2024; 15:1478263. [PMID: 39568995 PMCID: PMC11576456 DOI: 10.3389/fmicb.2024.1478263] [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: 08/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
Given the increasing concern about antimicrobial resistance among the microorganisms that cause infections in our society, there is an urgent need for new drug discovery. Currently, this process involves testing many low-quality compounds, resulting from the in vivo testing, on mammal models, which not only wastes time, resources, and money, but also raises ethical questions. In this review, we have discussed the potential of D. melanogaster as an intermediary experimental model in this drug discovery timeline. We have tackled the topic from a methodological perspective, providing recommendations regarding the range of drug concentrations to test based on the mechanism of action of each compound; how to treat D. melanogaster, how to monitor that treatment, and what parameters we should consider when designing a drug screening protocol to maximize the study's benefits. We also discuss the necessary improvements needed to establish the D. melanogaster model of infection as a standard technique in the drug screening process. Overall, D. melanogaster has been demonstrated to be a manageable model for studying broad-spectrum infection treatment. It allows us to obtain valuable information in a cost-effective manner, which can improve the drug screening process and provide insights into our current major concern. This approach is also in line with the 3R policy in biomedical research, in particular on the replacement and reduce the use of vertebrates in preclinical development.
Collapse
Affiliation(s)
- Maria Vidal
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Marta Arch
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Esther Fuentes
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Pere-Joan Cardona
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
12
|
Chasse AY, Bandyadka S, Wertheimer MC, Serizier SB, McCall K. Professional phagocytes are recruited for the clearance of obsolete nonprofessional phagocytes in the Drosophila ovary. Front Immunol 2024; 15:1389674. [PMID: 38994369 PMCID: PMC11236694 DOI: 10.3389/fimmu.2024.1389674] [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: 02/22/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Cell death is an important process in the body, as it occurs throughout every tissue during development, disease, and tissue regeneration. Phagocytes are responsible for clearing away dying cells and are typically characterized as either professional or nonprofessional phagocytes. Professional phagocytes, such as macrophages, are found in nearly every part of the body while nonprofessional phagocytes, such as epithelial cells, are found in every tissue type. However, there are organs that are considered "immune-privileged" as they have little to no immune surveillance and rely on nonprofessional phagocytes to engulf dying cells. These organs are surrounded by barriers to protect the tissue from viruses, bacteria, and perhaps even immune cells. The Drosophila ovary is considered immune-privileged, however the presence of hemocytes, the macrophages of Drosophila, around the ovary suggests they may have a potential function. Here we analyze hemocyte localization and potential functions in response to starvation-induced cell death in the ovary. Hemocytes were found to accumulate in the oviduct in the vicinity of mature eggs and follicle cell debris. Genetic ablation of hemocytes revealed that the presence of hemocytes affects oogenesis and that they phagocytose ovarian cell debris and in their absence fecundity decreases. Unpaired3, an IL-6 like cytokine, was found to be required for the recruitment of hemocytes to the oviduct to clear away obsolete follicle cells. These findings demonstrate a role for hemocytes in the ovary, providing a more thorough understanding of phagocyte communication and cell clearance in a previously thought immune-privileged organ.
Collapse
Affiliation(s)
- Alexandra Y. Chasse
- Program in Molecular Biology, Cell Biology & Biochemistry, Boston University, Boston, MA, United States
| | - Shruthi Bandyadka
- Program in Bioinformatics, Boston University, Boston, MA, United States
| | | | - Sandy B. Serizier
- Program in Molecular Biology, Cell Biology & Biochemistry, Boston University, Boston, MA, United States
| | - Kimberly McCall
- Department of Biology, Boston University, Boston, MA, United States
| |
Collapse
|
13
|
Wang J, Xue H, Yi X, Kim H, Hao Y, Jin LH. InR and Pi3K maintain intestinal homeostasis through STAT/EGFR and Notch signaling in enteroblasts. J Cell Biochem 2024; 125:e30545. [PMID: 38436545 DOI: 10.1002/jcb.30545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
To maintain the integrity of the adult gut, the proliferation and differentiation of stem cells must be strictly controlled. Several signaling pathways control the proliferation and differentiation of Drosophila intestinal epithelial cells. Although the modulatory effects of insulin pathway components on cell proliferation have been characterized, their specific role in which cell type and how these components interact with other regulatory signaling pathways remain largely unclear. In this study, we found that InR/Pi3K has major functions in enteroblasts (EBs) that were not previously described. The absence of InR/Pi3K in progenitors leads to a decrease in the number of EBs, while it has no significant effect on intestinal stem cells (ISCs). In addition, we found that InR/Pi3K regulates Notch activity in ISCs and EBs in an opposite way. This is also the reason for the decrease in EB. On the one hand, aberrantly low levels of Notch signaling in ISCs inhibit their proper differentiation into EBs; on the other hand, the higher Notch levels in EBs promote their excessive differentiation into enterocytes (ECs), leading to marked increases in abnormal ECs and decreased proliferation. Moreover, we found that Upd/JAK/STAT signaling acts as an effector or modifier of InR/Pi3K function in the midgut and cooperates with EGFR signaling to regulate cell proliferation. Altogether, our results demonstrate that InR and Pi3K are essential for coordinating stem cell differentiation and proliferation to maintain intestinal homeostasis.
Collapse
Affiliation(s)
- Jiewei Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Hongmei Xue
- Department of Children's Emergency Medicine, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xinyu Yi
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Hyonil Kim
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
- College of Life Science, Kim ll Sung University, Pyongyang, North Korea
| | - Yangguang Hao
- Department of Basic Medical, Shenyang Medical College, Shenyang, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
14
|
Liu Z, Zhang H, Lemaitre B, Li X. Duox activation in Drosophila Malpighian tubules stimulates intestinal epithelial renewal through a countercurrent flow. Cell Rep 2024; 43:114109. [PMID: 38613782 DOI: 10.1016/j.celrep.2024.114109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 02/26/2024] [Accepted: 03/28/2024] [Indexed: 04/15/2024] Open
Abstract
The gut must perform a dual role of protecting the host against toxins and pathogens while harboring mutualistic microbiota. Previous studies suggested that the NADPH oxidase Duox contributes to intestinal homeostasis in Drosophila by producing reactive oxygen species (ROS) in the gut that stimulate epithelial renewal. We find instead that the ROS generated by Duox in the Malpighian tubules leads to the production of Upd3, which enters the gut and stimulates stem cell proliferation. We describe in Drosophila the existence of a countercurrent flow system, which pushes tubule-derived Upd3 to the anterior part of the gut and stimulates epithelial renewal at a distance. Thus, our paper clarifies the role of Duox in gut homeostasis and describes the existence of retrograde fluid flow in the gut, collectively revealing a fascinating example of inter-organ communication.
Collapse
Affiliation(s)
- Zhonggeng Liu
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Hongyu Zhang
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Xiaoxue Li
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China; Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
15
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
16
|
Wang J, Li X, Wang X, Zhang C, Hao Y, Jin LH. The zinc finger protein CG12744 regulates intestinal stem cells in aged Drosophila through the EGFR and BMP pathways. Life Sci 2024; 340:122485. [PMID: 38311220 DOI: 10.1016/j.lfs.2024.122485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
AIM Aging is a process characterized by a time-dependent decline in the functionality of adult stem cells and is closely associated with age-related diseases. However, understanding how aging promotes disease and its underlying causes is critical for combating aging. MAIN METHODS The offspring of UAS-Gal4 and CG12744RNAiDrosophila were cultured for 33 days to evaluate the role of CG12744 in the aging intestine. Immunofluorescence was performed to detect specific cell type markers for assessing proliferation and differentiation. qRT-PCR was used to observe the changes in signaling regulating intestinal homeostasis in the aging intestine after CG12744 knockdown. 16S rRNA-seq analysis was also conducted to elucidate the role of gut microbes in CG12744-mediated intestinal dysfunction. KEY FINDINGS The mRNA levels of CG12744 were significantly increased in the aged midguts. Knockdown of CG12744 in progenitor cells further exacerbates the age-related intestinal hyperplasia and dysfunction. In particular, upon depletion of CG12744 in progenitors, enteroblasts (EBs) exhibited an increased propensity to differentiate along the enteroendocrine cell (EE) lineage. In contrast, the overexpression of CG12744 in progenitor cells restrained age-related gut hyperplasia in Drosophila. Moreover, CG12744 prevented age-related intestinal stem cell (ISC) overproliferation and differentiation by modulating the EGFR, JNK, and BMP pathways. In addition, the inhibition of CG12744 resulted in a significant increase in the gut microbial composition in aging flies. SIGNIFICANCE This study established a role for the CG12744 in regulating the proliferation and differentiation of adult stem cells, thereby identifying a potential therapeutic target for diseases caused by age-related dysfunction stem cell dysfunction.
Collapse
Affiliation(s)
- Jiewei Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin 150040, China
| | - Xianhao Li
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin 150040, China
| | - Xiaoran Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin 150040, China
| | - Chengcheng Zhang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin 150040, China
| | - Yangguang Hao
- Department of Basic Medical, Shenyang Medical College, Shenyang 110034, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No.26 Hexing Road Xiangfang District, Harbin 150040, China.
| |
Collapse
|
17
|
Awais MM, Fei S, Xia J, Feng M, Sun J. Insights into midgut cell types and their crucial role in antiviral immunity in the lepidopteran model Bombyx mori. Front Immunol 2024; 15:1349428. [PMID: 38420120 PMCID: PMC10899340 DOI: 10.3389/fimmu.2024.1349428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
The midgut, a vital component of the digestive system in arthropods, serves as an interface between ingested food and the insect's physiology, playing a pivotal role in nutrient absorption and immune defense mechanisms. Distinct cell types, including columnar, enteroendocrine, goblet and regenerative cells, comprise the midgut in insects and contribute to its robust immune response. Enterocytes/columnar cells, the primary absorptive cells, facilitate the immune response through enzyme secretions, while regenerative cells play a crucial role in maintaining midgut integrity by continuously replenishing damaged cells and maintaining the continuity of the immune defense. The peritrophic membrane is vital to the insect's innate immunity, shielding the midgut from pathogens and abrasive food particles. Midgut juice, a mixture of digestive enzymes and antimicrobial factors, further contributes to the insect's immune defense, helping the insect to combat invading pathogens and regulate the midgut microbial community. The cutting-edge single-cell transcriptomics also unveiled previously unrecognized subpopulations within the insect midgut cells and elucidated the striking similarities between the gastrointestinal tracts of insects and higher mammals. Understanding the intricate interplay between midgut cell types provides valuable insights into insect immunity. This review provides a solid foundation for unraveling the complex roles of the midgut, not only in digestion but also in immunity. Moreover, this review will discuss the novel immune strategies led by the midgut employed by insects to combat invading pathogens, ultimately contributing to the broader understanding of insect physiology and defense mechanisms.
Collapse
Affiliation(s)
| | | | | | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Hersperger F, Meyring T, Weber P, Chhatbar C, Monaco G, Dionne MS, Paeschke K, Prinz M, Groß O, Classen AK, Kierdorf K. DNA damage signaling in Drosophila macrophages modulates systemic cytokine levels in response to oxidative stress. eLife 2024; 12:RP86700. [PMID: 38189792 PMCID: PMC10945508 DOI: 10.7554/elife.86700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Environmental factors, infection, or injury can cause oxidative stress in diverse tissues and loss of tissue homeostasis. Effective stress response cascades, conserved from invertebrates to mammals, ensure reestablishment of homeostasis and tissue repair. Hemocytes, the Drosophila blood-like cells, rapidly respond to oxidative stress by immune activation. However, the precise signals how they sense oxidative stress and integrate these signals to modulate and balance the response to oxidative stress in the adult fly are ill-defined. Furthermore, hemocyte diversification was not explored yet on oxidative stress. Here, we employed high-throughput single nuclei RNA-sequencing to explore hemocytes and other cell types, such as fat body, during oxidative stress in the adult fly. We identified distinct cellular responder states in plasmatocytes, the Drosophila macrophages, associated with immune response and metabolic activation upon oxidative stress. We further define oxidative stress-induced DNA damage signaling as a key sensor and a rate-limiting step in immune-activated plasmatocytes controlling JNK-mediated release of the pro-inflammatory cytokine unpaired-3. We subsequently tested the role of this specific immune activated cell stage during oxidative stress and found that inhibition of DNA damage signaling in plasmatocytes, as well as JNK or upd3 overactivation, result in a higher susceptibility to oxidative stress. Our findings uncover that a balanced composition and response of hemocyte subclusters is essential for the survival of adult Drosophila on oxidative stress by regulating systemic cytokine levels and cross-talk to other organs, such as the fat body, to control energy mobilization.
Collapse
Affiliation(s)
- Fabian Hersperger
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
| | - Tim Meyring
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Pia Weber
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Chintan Chhatbar
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Gianni Monaco
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of FreiburgFreiburgGermany
| | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Katrin Paeschke
- Department of Oncology, Haematology and Rheumatology, University Hospital BonnBonnGermany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital BonnBonnGermany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- Signalling Research Centres BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- Signalling Research Centres BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, Faculty of Biology, University of FreiburgFreiburgGermany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| |
Collapse
|
19
|
Christensen CF, Laurichesse Q, Loudhaief R, Colombani J, Andersen DS. Drosophila activins adapt gut size to food intake and promote regenerative growth. Nat Commun 2024; 15:273. [PMID: 38177201 PMCID: PMC10767106 DOI: 10.1038/s41467-023-44553-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
Rapidly renewable tissues adapt different strategies to cope with environmental insults. While tissue repair is associated with increased intestinal stem cell (ISC) proliferation and accelerated tissue turnover rates, reduced calorie intake triggers a homeostasis-breaking process causing adaptive resizing of the gut. Here we show that activins are key drivers of both adaptive and regenerative growth. Activin-β (Actβ) is produced by stem and progenitor cells in response to intestinal infections and stimulates ISC proliferation and turnover rates to promote tissue repair. Dawdle (Daw), a divergent Drosophila activin, signals through its receptor, Baboon, in progenitor cells to promote their maturation into enterocytes (ECs). Daw is dynamically regulated during starvation-refeeding cycles, where it couples nutrient intake with progenitor maturation and adaptive resizing of the gut. Our results highlight an activin-dependent mechanism coupling nutrient intake with progenitor-to-EC maturation to promote adaptive resizing of the gut and further establish activins as key regulators of adult tissue plasticity.
Collapse
Affiliation(s)
- Christian F Christensen
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, 2100 Copenhagen O, Copenhagen, Denmark
| | - Quentin Laurichesse
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, 2100 Copenhagen O, Copenhagen, Denmark
| | - Rihab Loudhaief
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, 2100 Copenhagen O, Copenhagen, Denmark
| | - Julien Colombani
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, 2100 Copenhagen O, Copenhagen, Denmark.
| | - Ditte S Andersen
- Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 15, Build. 3, 3rd floor, 2100 Copenhagen O, Copenhagen, Denmark.
| |
Collapse
|
20
|
Prakash A, Monteith KM, Bonnet M, Vale PF. Duox and Jak/Stat signalling influence disease tolerance in Drosophila during Pseudomonas entomophila infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104756. [PMID: 37302730 DOI: 10.1016/j.dci.2023.104756] [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: 04/26/2023] [Revised: 05/18/2023] [Accepted: 06/09/2023] [Indexed: 06/13/2023]
Abstract
Disease tolerance describes an infected host's ability to maintain health independently of the ability to clear microbe loads. The Jak/Stat pathway plays a pivotal role in humoral innate immunity by detecting tissue damage and triggering cellular renewal, making it a candidate tolerance mechanism. Here, we find that in Drosophila melanogaster infected with Pseudomonas entomophila disrupting ROS-producing dual oxidase (duox) or the negative regulator of Jak/Stat Socs36E, render male flies less tolerant. Another negative regulator of Jak/Stat, G9a - which has previously been associated with variable tolerance of viral infections - did not affect the rate of mortality with increasing microbe loads compared to flies with functional G9a, suggesting it does not affect tolerance of bacterial infection as in viral infection. Our findings highlight that ROS production and Jak/Stat signalling influence the ability of flies to tolerate bacterial infection sex-specifically and may therefore contribute to sexually dimorphic infection outcomes in Drosophila.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK.
| | - Katy M Monteith
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK
| | - Mickael Bonnet
- UFR De Biologie, Campus Universitaire Des Cezeaux, France
| | - Pedro F Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK.
| |
Collapse
|
21
|
Arch M, Vidal M, Fuentes E, Abat AS, Cardona PJ. The reproductive status determines tolerance and resistance to Mycobacterium marinum in Drosophila melanogaster. Evol Med Public Health 2023; 11:332-347. [PMID: 37868078 PMCID: PMC10590161 DOI: 10.1093/emph/eoad029] [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: 01/14/2023] [Revised: 07/27/2023] [Indexed: 10/24/2023] Open
Abstract
Sex and reproductive status of the host have a major impact on the immune response against infection. Our aim was to understand their impact on host tolerance or resistance in the systemic Mycobacterium marinum infection of Drosophila melanogaster. We measured host survival and bacillary load at time of death, as well as expression by quantitative real-time polymerase chain reaction of immune genes (diptericin and drosomycin). We also assessed the impact of metabolic and hormonal regulation in the protection against infection by measuring expression of upd3, impl2 and ecR. Our data showed increased resistance in actively mating flies and in mated females, while reducing their tolerance to infection. Data suggests that Toll and immune deficiency (Imd) pathways determine tolerance and resistance, respectively, while higher basal levels of ecR favours the stimulation of the Imd pathway. A dual role has been found for upd3 expression, linked to increased/decreased mycobacterial load at the beginning and later in infection, respectively. Finally, impl2 expression has been related to increased resistance in non-actively mating males. These results allow further assessment on the differences between sexes and highlights the role of the reproductive status in D. melanogaster to face infections, demonstrating their importance to determine resistance and tolerance against M. marinum infection.
Collapse
Affiliation(s)
- Marta Arch
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
| | - Maria Vidal
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Esther Fuentes
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
| | - Anmaw Shite Abat
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Department of Veterinary Paraclinical Studies, University of Gondar, Gondar, Ethiopia
| | - Pere-Joan Cardona
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
22
|
Zhou J, Boutros M. Intestinal stem cells and their niches in homeostasis and disease. Cells Dev 2023; 175:203862. [PMID: 37271243 DOI: 10.1016/j.cdev.2023.203862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/21/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Tissues such as the intestine harbor stem cells that have remarkable functional plasticity in response to a dynamic environment. To adapt to the environment, stem cells constantly receive information from their surrounding microenvironment (also called the 'niche') that instructs them how to adapt to changes. The Drosophila midgut shows morphological and functional similarities to the mammalian small intestine and has been a useful model system to study signaling events in stem cells and tissue homeostasis. In this review, we summarize the current understanding of the Drosophila midgut regarding how stem cells communicate with microenvironmental niches including enteroblasts, enterocytes, enteroendocrine cells and visceral muscles to coordinate tissue regeneration and homeostasis. In addition, distant cells such as hemocytes or tracheal cells have been shown to interact with stem cells and influence the development of intestinal diseases. We discuss the contribution of stem cell niches in driving or counteracting disease progression, and review conceptual advances derived from the Drosophila intestine as a model for stem cell biology.
Collapse
Affiliation(s)
- Jun Zhou
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany; School of Biomedical Sciences, Hunan University, Changsha, China.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| |
Collapse
|
23
|
Kuyateh O, Obbard DJ. Viruses in Laboratory Drosophila and Their Impact on Host Gene Expression. Viruses 2023; 15:1849. [PMID: 37766256 PMCID: PMC10537266 DOI: 10.3390/v15091849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Drosophila melanogaster has one of the best characterized antiviral immune responses among invertebrates. However, relatively few easily transmitted natural virus isolates are available, and so many Drosophila experiments have been performed using artificial infection routes and artificial host-virus combinations. These may not reflect natural infections, especially for subtle phenotypes such as gene expression. Here, to explore the laboratory virus community and to better understand how natural virus infections induce changes in gene expression, we have analysed seven publicly available D. melanogaster transcriptomic sequencing datasets that were originally sequenced for projects unrelated to virus infection. We have found ten known viruses-including five that have not been experimentally isolated-but no previously unknown viruses. Our analysis of host gene expression revealed that numerous genes were differentially expressed in flies that were naturally infected with a virus. For example, flies infected with nora virus showed patterns of gene expression consistent with intestinal vacuolization and possible host repair via the upd3 JAK/STAT pathway. We also found marked sex differences in virus-induced differential gene expression. Our results show that natural virus infection in laboratory Drosophila does indeed induce detectable changes in gene expression, suggesting that this may form an important background condition for experimental studies in the laboratory.
Collapse
Affiliation(s)
- Oumie Kuyateh
- Institute of Ecology and Evolution, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK;
- Parasites and Microbes, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Darren J. Obbard
- Institute of Ecology and Evolution, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK;
| |
Collapse
|
24
|
Petsakou A, Liu Y, Liu Y, Comjean A, Hu Y, Perrimon N. Epithelial Ca 2+ waves triggered by enteric neurons heal the gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553227. [PMID: 37645990 PMCID: PMC10461974 DOI: 10.1101/2023.08.14.553227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the etiology of chronic disorders such as inflammatory bowel diseases and cancer. We used the Drosophila midgut to investigate this question and discovered that during regeneration a subpopulation of cholinergic enteric neurons triggers Ca2+ currents among enterocytes to promote return of the epithelium to homeostasis. Specifically, we found that down-regulation of the cholinergic enzyme Acetylcholinesterase in the epithelium enables acetylcholine from defined enteric neurons, referred as ARCENs, to activate nicotinic receptors in enterocytes found near ARCEN-innervations. This activation triggers high Ca2+ influx that spreads in the epithelium through Inx2/Inx7 gap junctions promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki activation and increase of inflammatory cytokines together with hyperplasia, reminiscent of inflammatory bowel diseases. Altogether, we found that during gut regeneration the conserved cholinergic pathway facilitates epithelial Ca2+ waves that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric-dependent intestinal regeneration which advance the current understanding of how a tissue returns to its homeostatic state after injury and could ultimately help existing therapeutics.
Collapse
Affiliation(s)
| | - Yifang Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Aram Comjean
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Boston, USA
| |
Collapse
|
25
|
Tafesh-Edwards G, Eleftherianos I. Functional role of thioester-containing proteins in the Drosophila anti-pathogen immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104578. [PMID: 36270515 DOI: 10.1016/j.dci.2022.104578] [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: 07/08/2022] [Revised: 09/17/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Thioester-containing proteins (TEPs) are present in many animal species ranging from deuterostomes to protostomes, which emphasizes their evolutionary conservation and importance in animal physiology. Phylogenetically, insect TEPs share sequence similarity with mammalian α2-macroglobulin. Drosophila melanogaster is specifically considered a superb model for teasing apart innate immune processes. Here we review recent discoveries on the involvement of Drosophila TEPs in the immune response against bacterial pathogens, nematode parasites, and parasitoid wasps. This information generates novel insights into the role of TEPs as regulators of homeostasis in Drosophila and supports the complexity of immune recognition and specificity in insects and more generally in invertebrates. These developments together with recent advances in gene editing and multi-omics will enable the fly immunity community to appreciate the molecular and mechanistic contributions of TEPs to the modulation of the host defense against infectious disease and possibly to translate this information into tangible therapeutic benefits.
Collapse
Affiliation(s)
- Ghada Tafesh-Edwards
- 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
|
26
|
Zeng T, Jaffar S, Xu Y, Qi Y. The Intestinal Immune Defense System in Insects. Int J Mol Sci 2022; 23:ijms232315132. [PMID: 36499457 PMCID: PMC9740067 DOI: 10.3390/ijms232315132] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Over a long period of evolution, insects have developed unique intestinal defenses against invasion by foreign microorganisms, including physical defenses and immune responses. The physical defenses of the insect gut consist mainly of the peritrophic matrix (PM) and mucus layer, which are the first barriers to pathogens. Gut microbes also prevent the colonization of pathogens. Importantly, the immune-deficiency (Imd) pathways produce antimicrobial peptides to eliminate pathogens; mechanisms related to reactive oxygen species are another important pathway for insect intestinal immunity. The janus kinase/STAT signaling pathway is involved in intestinal immunity by producing bactericidal substances and regulating tissue repair. Melanization can produce many bactericidal active substances into the intestine; meanwhile, there are multiple responses in the intestine to fight against viral and parasitic infections. Furthermore, intestinal stem cells (ISCs) are also indispensable in intestinal immunity. Only the coordinated combination of the intestinal immune defense system and intestinal tissue renewal can effectively defend against pathogenic microorganisms.
Collapse
|
27
|
Keshav N, Ammankallu R, Shashidhar, Paithankar JG, Baliga MS, Patil RK, Kudva AK, Raghu SV. Dextran sodium sulfate alters antioxidant status in the gut affecting the survival of Drosophila melanogaster. 3 Biotech 2022; 12:280. [PMID: 36275361 PMCID: PMC9481858 DOI: 10.1007/s13205-022-03349-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of disorders characterized by chronic inflammation in the intestine. Several studies confirmed that oxidative stress induced by an enormous amount of reactive free radicals triggers the onset of IBD. Currently, there is an increasing trend in the global incidence of IBD and it is coupled with a lack of adequate long-term therapeutic options. At the same time, progress in research to understand the pathogenesis of IBD has been hampered due to the absence of adequate animal models. Currently, the toxic chemical Dextran Sulfate Sodium (DSS) induced gut inflammation in rodents is widely perceived as a good model of experimental colitis or IBD. Drosophila melanogaster, a genetic animal model, shares ~ 75% sequence similarity to genes causing different diseases in humans and also has conserved digestion and absorption features. Therefore, in the current study, we used Drosophila as a model system to induce and investigate DSS-induced colitis. Anatomical, biochemical, and molecular analyses were performed to measure the levels of inflammation and cellular disturbances in the gastrointestinal (GI) tract of Drosophila. Our study shows that DSS-induced inflammation lowers the levels of antioxidant molecules, affects the life span, reduces physiological activity and induces cellular damage in the GI tract mimicking pathophysiological features of IBD in Drosophila. Such a DSS-induced Drosophila colitis model can be further used for understanding the molecular pathology of IBD and screening novel drugs. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03349-2.
Collapse
Affiliation(s)
- Nishal Keshav
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Karnataka India
| | - Ramyalakshmi Ammankallu
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Karnataka India
| | - Shashidhar
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Karnataka India
| | - Jagdish Gopal Paithankar
- Nitte University Center for Science Education and Research (NUCSER), Nitte (Deemed to be University), Mangalore, 575018 India
| | | | - Rajashekhar K. Patil
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Karnataka India
| | - Avinash Kundadka Kudva
- Department of Biochemistry, Mangalore University, Mangalagangothri, 574199 Karnataka India
| | - Shamprasad Varija Raghu
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Karnataka India
| |
Collapse
|
28
|
Kausar S, Gul I, Liu R, Ke XX, Dong Z, Abbas MN, Cui H. Antheraea pernyi Suppressor of Cytokine Signaling 2 Negatively Modulates the JAK/STAT Pathway to Attenuate Microbial Infection. Int J Mol Sci 2022; 23:ijms231810389. [PMID: 36142300 PMCID: PMC9499667 DOI: 10.3390/ijms231810389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (STAT) pathway has been shown to govern various physiological processes, including immune responses, hematopoiesis, cell growth, and differentiation. Recent studies show that suppressors of cytokine signaling (SOCS) proteins attenuate JAK-STAT signaling in mammals; however, their functions are less clear in lepidopteran insects. Here, we report a full-length sequence of SOCS-2 from the Chinese oak silkworm Antheraea pernyi (designated as ApSOCS-2) and study its biological role in immune responses via the JAK-STAT pathway. ApSOCS-2 expression was high in the fat bodies and hemocytes of A. pernyi fifth instar larvae. After pathogen infection with nucleopolyhedrovirus, Beauveria bassiana, Escherichia coli, and Microccus luteus, ApSOCS-2 mRNA was strongly increased compared to the control group. To elucidate the possible involvement in innate immunity, we measured antimicrobial peptide genes expression profiles in the fat body of A. pernyi. In contrast, recombinant ApSOCS-2 protein administration significantly reduced the AMPs transcription, while the depletion of ApSOCS-2 by RNAi increased their expression. Furthermore, we observed higher antibacterial activity and lower bacterial replication in dsApSOCS-2-treated larvae. The ApSOCS-2 transcription level was reduced in STAT depleted A. pernyi larvae challenged by M. luteus. The ApSOCS-2 RNAi data sets were also subjected to transcriptomic analysis, which suggests that ApSOCS-2 is a key regulator of immune function. Taken together, our data suggest that ApSOCS-2 is required for the negative regulation of AMPs transcripts via the JAK-STAT pathway in the insect.
Collapse
Affiliation(s)
- Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Isma Gul
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Ruochen Liu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Xiao-Xue Ke
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (M.N.A.); (H.C.); Tel.: +86-23-68251712 (H.C.)
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing 400716, China
- Affiliation Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (M.N.A.); (H.C.); Tel.: +86-23-68251712 (H.C.)
| |
Collapse
|
29
|
Yan Y, Sigle LT, Rinker DC, Estévez-Lao TY, Capra JA, Hillyer JF. The immune deficiency and c-Jun N-terminal kinase pathways drive the functional integration of the immune and circulatory systems of mosquitoes. Open Biol 2022; 12:220111. [PMID: 36069078 PMCID: PMC9449813 DOI: 10.1098/rsob.220111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune and circulatory systems of animals are functionally integrated. In mammals, the spleen and lymph nodes filter and destroy microbes circulating in the blood and lymph, respectively. In insects, immune cells that surround the heart valves (ostia), called periostial haemocytes, destroy pathogens in the areas of the body that experience the swiftest haemolymph (blood) flow. An infection recruits additional periostial haemocytes, amplifying heart-associated immune responses. Although the structural mechanics of periostial haemocyte aggregation have been defined, the genetic factors that regulate this process remain less understood. Here, we conducted RNA sequencing in the African malaria mosquito, Anopheles gambiae, and discovered that an infection upregulates multiple components of the immune deficiency (IMD) and c-Jun N-terminal kinase (JNK) pathways in the heart with periostial haemocytes. This upregulation is greater in the heart with periostial haemocytes than in the circulating haemocytes or the entire abdomen. RNA interference-based knockdown then showed that the IMD and JNK pathways drive periostial haemocyte aggregation and alter phagocytosis and melanization on the heart, thereby demonstrating that these pathways regulate the functional integration between the immune and circulatory systems. Understanding how insects fight infection lays the foundation for novel strategies that could protect beneficial insects and harm detrimental ones.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Leah T. Sigle
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - David C. Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - John A. Capra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA,Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
30
|
Prakash A, Monteith KM, Vale PF. Mechanisms of damage prevention, signalling and repair impact disease tolerance. Proc Biol Sci 2022; 289:20220837. [PMID: 35975433 PMCID: PMC9382215 DOI: 10.1098/rspb.2022.0837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The insect gut is frequently exposed to pathogenic threats and must not only clear these potential infections, but also tolerate relatively high microbe loads. In contrast to the mechanisms that eliminate pathogens, we currently know less about the mechanisms of disease tolerance. We investigated how well-described mechanisms that prevent, signal, control or repair damage during infection contribute to the phenotype of disease tolerance. We established enteric infections with the bacterial pathogen Pseudomonas entomophila in transgenic lines of Drosophila melanogaster fruit flies affecting dcy (a major component of the peritrophic matrix), upd3 (a cytokine-like molecule), irc (a negative regulator of reactive oxygen species) and egfr1 (epithelial growth factor receptor). Flies lacking dcy experienced the highest mortality, while loss of function of either irc or upd3 reduced tolerance in both sexes. The disruption of egfr1 resulted in a severe loss in tolerance in male flies but had no substantial effect on the ability of female flies to tolerate P. entomophila infection, despite carrying greater microbe loads than males. Together, our findings provide evidence for the role of damage limitation mechanisms in disease tolerance and highlight how sexual dimorphism in these mechanisms could generate sex differences in infection outcomes.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Katy M. Monteith
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Pedro F. Vale
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| |
Collapse
|
31
|
Yang S, Li X, Xiu M, Dai Y, Wan S, Shi Y, Liu Y, He J. Flos puerariae ameliorates the intestinal inflammation of Drosophila via modulating the Nrf2/Keap1, JAK-STAT and Wnt signaling. Front Pharmacol 2022; 13:893758. [PMID: 36059974 PMCID: PMC9432424 DOI: 10.3389/fphar.2022.893758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
Gut homeostasis is important for human health, and its disruption can lead to inflammatory bowel disease (IBD). Flos Puerariae is a herb with a wide variety of pharmacological activities including antioxidant, antidiabetic, antialcoholismic and anti-inflammatory properties. However, the role of Flos Puerariae on treating IBD remains obscure. Here, we employed Drosophila melanogaster as a model organism to investigate the protective effect of Flos Puerariae extract (FPE) against sodium dodecyl sulfate (SDS)-induced intestinal injury. Our data showed that FPE had no toxic effect in flies, and significantly extended lifespan in SDS-inflamed flies, reduced stem cell proliferation in the midgut, and maintained intestinal morphological integrity. Furthermore, FPE remarkably recused the altered expression level of genes and proteins in Nrf2/Keap1 signaling, JAK-STAT signaling and Wnt signaling pathways in gut of inflammation flies. Thus, FPE has a protective effect against intestinal injury possibly via increasing the Nrf2/keap1 pathway and suppressing the JAK-STAT and Wnt signaling pathways, which would have tremendous potential for treating IBD.
Collapse
Affiliation(s)
- Shipei Yang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xu Li
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Minghui Xiu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
- Research Center of Traditional Chinese Medicine in Gansu, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuting Dai
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shengfang Wan
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Shi
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
- *Correspondence: Jianzheng He, ; Yongqi Liu,
| | - Jianzheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
- *Correspondence: Jianzheng He, ; Yongqi Liu,
| |
Collapse
|
32
|
Yu S, Luo F, Xu Y, Zhang Y, Jin LH. Drosophila Innate Immunity Involves Multiple Signaling Pathways and Coordinated Communication Between Different Tissues. Front Immunol 2022; 13:905370. [PMID: 35911716 PMCID: PMC9336466 DOI: 10.3389/fimmu.2022.905370] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune response provides the first line of defense against invading pathogens, and immune disorders cause a variety of diseases. The fruit fly Drosophila melanogaster employs multiple innate immune reactions to resist infection. First, epithelial tissues function as physical barriers to prevent pathogen invasion. In addition, macrophage-like plasmatocytes eliminate intruders through phagocytosis, and lamellocytes encapsulate large particles, such as wasp eggs, that cannot be phagocytosed. Regarding humoral immune responses, the fat body, equivalent to the mammalian liver, secretes antimicrobial peptides into hemolymph, killing bacteria and fungi. Drosophila has been shown to be a powerful in vivo model for studying the mechanism of innate immunity and host-pathogen interactions because Drosophila and higher organisms share conserved signaling pathways and factors. Moreover, the ease with which Drosophila genetic and physiological characteristics can be manipulated prevents interference by adaptive immunity. In this review, we discuss the signaling pathways activated in Drosophila innate immunity, namely, the Toll, Imd, JNK, JAK/STAT pathways, and other factors, as well as relevant regulatory networks. We also review the mechanisms by which different tissues, including hemocytes, the fat body, the lymph gland, muscles, the gut and the brain coordinate innate immune responses. Furthermore, the latest studies in this field are outlined in this review. In summary, understanding the mechanism underlying innate immunity orchestration in Drosophila will help us better study human innate immunity-related diseases.
Collapse
|
33
|
Abstract
Adult tissues in Metazoa dynamically remodel their structures in response to environmental challenges including sudden injury, pathogen infection, and nutritional fluctuation, while maintaining quiescence under homoeostatic conditions. This characteristic, hereafter referred to as adult tissue plasticity, can prevent tissue dysfunction and improve the fitness of organisms in continuous and/or severe change of environments. With its relatively simple tissue structures and genetic tools, studies using the fruit fly Drosophila melanogaster have provided insights into molecular mechanisms that control cellular responses, particularly during regeneration and nutrient adaptation. In this review, we present the current understanding of cellular mechanisms, stem cell proliferation, polyploidization, and cell fate plasticity, all of which enable adult tissue plasticity in various Drosophila adult organs including the midgut, the brain, and the gonad, and discuss the organismal strategy in response to environmental changes and future directions of the research.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Yu-Ichiro Nakajima
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Xu DC, Wang L, Yamada KM, Baena-Lopez LA. Non-apoptotic activation of Drosophila caspase-2/9 modulates JNK signaling, the tumor microenvironment, and growth of wound-like tumors. Cell Rep 2022; 39:110718. [PMID: 35443185 PMCID: PMC9082238 DOI: 10.1016/j.celrep.2022.110718] [Citation(s) in RCA: 4] [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: 08/04/2020] [Revised: 02/15/2022] [Accepted: 03/31/2022] [Indexed: 11/23/2022] Open
Abstract
Resistance to apoptosis due to caspase deregulation is considered one of the main hallmarks of cancer. However, the discovery of novel non-apoptotic caspase functions has revealed unknown intricacies about the interplay between these enzymes and tumor progression. To investigate this biological problem, we capitalized on a Drosophila tumor model with human relevance based on the simultaneous overactivation of the EGFR and the JAK/STAT signaling pathways. Our data indicate that widespread non-apoptotic activation of initiator caspases limits JNK signaling and facilitates cell fate commitment in these tumors, thus preventing the overgrowth and exacerbation of malignant features of transformed cells. Intriguingly, caspase activity also reduces the presence of macrophage-like cells with tumor-promoting properties in the tumor microenvironment. These findings assign tumor-suppressing activities to caspases independent of apoptosis, while providing molecular details to better understand the contribution of these enzymes to tumor progression.
Collapse
Affiliation(s)
- Derek Cui Xu
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA; Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire OX1 3RE, UK
| | - Li Wang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire OX1 3RE, UK
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA.
| | | |
Collapse
|
35
|
Arinda BN, Innabi YA, Grasis JA, Oviedo NJ. Non-traditional roles of immune cells in regeneration: an evolutionary perspective. Development 2022; 149:275269. [PMID: 35502784 PMCID: PMC9124569 DOI: 10.1242/dev.199903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Immune cells are known to engage in pathogen defense. However, emerging research has revealed additional roles for immune cells, which are independent of their function in the immune response. Here, we underscore the ability of cells outside of the adaptive immune system to respond to recurring infections through the lens of evolution and cellular memory. With this in mind, we then discuss the bidirectional crosstalk between the immune cells and stem cells and present examples where these interactions regulate tissue repair and regeneration. We conclude by suggesting that comprehensive analyses of the immune system may enable biomedical applications in stem cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Beryl N Arinda
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Yacoub A Innabi
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Juris A Grasis
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA.,Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA.,Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| |
Collapse
|
36
|
Caravello G, Franchet A, Niehus S, Ferrandon D. Phagocytosis Is the Sole Arm of Drosophila melanogaster Known Host Defenses That Provides Some Protection Against Microsporidia Infection. Front Immunol 2022; 13:858360. [PMID: 35493511 PMCID: PMC9043853 DOI: 10.3389/fimmu.2022.858360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Microsporidia are obligate intracellular parasites able to infest specifically a large range of species, including insects. The knowledge about the biology of microsporidial infections remains confined to mostly descriptive studies, including molecular approaches such as transcriptomics or proteomics. Thus, functional data to understand insect host defenses are currently lacking. Here, we have undertaken a genetic analysis of known host defenses of the Drosophila melanogaster using an infection model whereby Tubulinosema ratisbonensis spores are directly injected in this insect. We find that phagocytosis does confer some protection in this infection model. In contrast, the systemic immune response, extracellular reactive oxygen species, thioester proteins, xenophagy, and intracellular antiviral response pathways do not appear to be involved in the resistance against this parasite. Unexpectedly, several genes such as PGRP-LE seem to promote this infection. The prophenol oxidases that mediate melanization have different functions; PPO1 presents a phenotype similar to that of PGRP-LE whereas that of PPO2 suggests a function in the resilience to infection. Similarly, eiger and Unpaired3, which encode two cytokines secreted by hemocytes display a resilience phenotype with a strong susceptibility to T. ratisbonensis.
Collapse
Affiliation(s)
| | | | | | - Dominique Ferrandon
- UPR9022, University of Strasbourg, Institut de Biologie Moléculaire et Cellulaire (IBMC), Modèles Insectes D’Immunité Innée (M3I) Unité Propre Recherche (UPR) 9022 du Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| |
Collapse
|
37
|
Abstract
Inflammatory response in Drosophila to sterile (axenic) injury in embryos and adults has received some attention in recent years, and most concentrate on the events at the injury site. Here we focus on the effect sterile injury has on the hematopoietic organ, the lymph gland, and the circulating blood cells in the larva, the developmental stage at which major events of hematopoiesis are evident. In mammals, injury activates Toll-like receptor/NF-κB signaling in macrophages, which then express and secrete secondary, proinflammatory cytokines. In Drosophila larvae, distal puncture injury of the body wall epidermis causes a rapid activation of Toll and Jun kinase (JNK) signaling throughout the hematopoietic system and the differentiation of a unique blood cell type, the lamellocyte. Furthermore, we find that Toll and JNK signaling are coupled in their activation. Secondary to this Toll/JNK response, a cytokine, Upd3, is induced as a Toll pathway transcriptional target, which then promotes JAK/STAT signaling within the blood cells. Toll and JAK/STAT signaling are required for the emergence of the injury-induced lamellocytes. This is akin to the derivation of specialized macrophages in mammalian systems. Upstream, at the injury site, a Duox- and peroxide-dependent signal causes the activation of the proteases Grass and SPE, needed for the activation of the Toll-ligand Spz, but microbial sensors or the proteases most closely associated with them during septic injury are not involved in the axenic inflammatory response.
Collapse
|
38
|
Domesticated LTR-Retrotransposon gag-Related Gene (Gagr) as a Member of the Stress Response Network in Drosophila. Life (Basel) 2022; 12:life12030364. [PMID: 35330115 PMCID: PMC8956099 DOI: 10.3390/life12030364] [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: 01/29/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/24/2022] Open
Abstract
The most important sources of new components of genomes are transposable elements, which can occupy more than half of the nucleotide sequence of the genome in higher eukaryotes. Among the mobile components of a genome, a special place is occupied by retroelements, which are similar to retroviruses in terms of their mechanisms of integration into a host genome. The process of positive selection of certain sequences of transposable elements and retroviruses in a host genome is commonly called molecular domestication. There are many examples of evolutionary adaptations of gag (retroviral capsid) sequences as new regulatory sequences of different genes in mammals, where domesticated gag genes take part in placenta functioning and embryogenesis, regulation of apoptosis, hematopoiesis, and metabolism. The only gag-related gene has been found in the Drosophila genome—Gagr. According to the large-scale transcriptomic and proteomic analysis data, the Gagr gene in D. melanogaster is a component of the protein complex involved in the stress response. In this work, we consider the evolutionary processes that led to the formation of a new function of the domesticated gag gene and its adaptation to participation in the stress response. We discuss the possible functional role of the Gagr as part of the complex with its partners in Drosophila, and the pathway of evolution of proteins of the complex in eukaryotes to determine the benefit of the domesticated retroelement gag gene.
Collapse
|
39
|
Soory A, Ratnaparkhi GS. SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 2022; 18:e1010356. [PMID: 35255103 PMCID: PMC8929699 DOI: 10.1371/journal.ppat.1010356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 03/17/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Post-translational modification by the small ubiquitin-like modifier, SUMO can modulate the activity of its conjugated proteins in a plethora of cellular contexts. The effect of SUMO conjugation of proteins during an immune response is poorly understood in Drosophila. We have previously identified that the transcription factor Jra, the Drosophila Jun ortholog and a member of the AP-1 complex is one such SUMO target. Here, we find that Jra is a regulator of the Pseudomonas entomophila induced gut immune gene regulatory network, modulating the expression of a few thousand genes, as measured by quantitative RNA sequencing. Decrease in Jra in gut enterocytes is protective, suggesting that reduction of Jra signaling favors the host over the pathogen. In Jra, lysines 29 and 190 are SUMO conjugation targets, with the JraK29R+K190R double mutant being SUMO conjugation resistant (SCR). Interestingly, a JraSCR fly line, generated by CRISPR/Cas9 based genome editing, is more sensitive to infection, with adults showing a weakened host response and increased proliferation of Pseudomonas. Transcriptome analysis of the guts of JraSCR and JraWT flies suggests that lack of SUMOylation of Jra significantly changes core elements of the immune gene regulatory network, which include antimicrobial agents, secreted ligands, feedback regulators, and transcription factors. Mechanistically, SUMOylation attenuates Jra activity, with the TFs, forkhead, anterior open, activating transcription factor 3 and the master immune regulator Relish being important transcriptional targets. Our study implicates Jra as a major immune regulator, with dynamic SUMO conjugation/deconjugation of Jra modulating the kinetics of the gut immune response. The intestine has a resident population of commensal microorganisms against which the immune machinery is tuned to show low or no reactivity. In contrast, when pathogenic microorganisms are ingested, the gut responds by activating signaling cascades that lead to the killing and clearance of the pathogen. In this study, we examine the role played by the well-known transcription factor Jun in regulating the immune response in the Drosophila gut. We find that loss of Jun leads to the change in intensity and kinetics of the gut immune transcriptome. The transcriptional profile indicates a stronger response when Jun activity is reduced. Also, animals infected with Pseudomonas entomophila live longer when Jun signaling is reduced. Further, we find that Jun is post-translationally modified on Lys29 and Lys190 by SUMO. To understand the effect of SUMO-conjugation of Jun, we create by state-of-the-art CRISPR/Cas9 genome editing a Drosophila line where Jun is resistant to SUMOylation. This line is more sensitive to infection, with a weaker host-defense response. Our data suggest that Jun Signaling favors the pathogen by dampening the immune response. SUMO conjugation of Jun reverses the dampening and strengthens the immune response in favor of the host. Dynamic SUMOylation of Jun thus fine-tunes the gut immune response to pathogens.
Collapse
Affiliation(s)
- Amarendranath Soory
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| | - Girish S. Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| |
Collapse
|
40
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
41
|
Gera J, Budakoti P, Suhag M, Mandal L, Mandal S. Physiological ROS controls Upd3-dependent modeling of ECM to support cardiac function in Drosophila. SCIENCE ADVANCES 2022; 8:eabj4991. [PMID: 35179958 PMCID: PMC8856619 DOI: 10.1126/sciadv.abj4991] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Despite their highly reactive nature, reactive oxygen species (ROS) at the physiological level serve as signaling molecules regulating diverse biological processes. While ROS usually act autonomously, they also function as local paracrine signals by diffusing out of the cells producing them. Using in vivo molecular genetic analyses in Drosophila, we provide evidence for ROS-dependent paracrine signaling that does not entail ROS release. We show that elevated levels of physiological ROS within the pericardial cells activate a signaling cascade transduced by Ask1, c-Jun N-terminal kinase, and p38 to regulate the expression of the cytokine Unpaired 3 (Upd3). Upd3 released by the pericardial cells controls fat body-specific expression of the extracellular matrix (ECM) protein Pericardin, essential for cardiac function and healthy life span. Therefore, our work reveals an unexpected inter-organ communication circuitry wherein high physiological levels of ROS regulate cytokine-dependent modulation of cardiac ECM with implications in normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Jayati Gera
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Prerna Budakoti
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Meghna Suhag
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab 140306, India
- Corresponding author.
| |
Collapse
|
42
|
Abstract
In adult insects, as in vertebrates, the gut epithelium is a highly regenerative tissue that can renew itself rapidly in response to changing inputs from nutrition, the gut microbiota, ingested toxins, and signals from other organs. Because of its cellular and genetic similarities to the mammalian intestine, and its relevance as a target for the control of insect pests and disease vectors, many researchers have used insect intestines to address fundamental questions about stem cell functions during tissue maintenance and regeneration. In Drosophila, where most of the experimental work has been performed, not only are intestinal cell types and behaviors well characterized, but numerous cell signaling interactions have been detailed that mediate gut epithelial regeneration. A prevailing model for regenerative responses in the insect gut invokes stress sensing by damaged enterocytes (ECs) as a principal source for signaling that activates the division of intestinal stem cells (ISCs) and the growth and differentiation of their progeny. However, extant data also reveal alternative mechanisms for regeneration that involve ISC-intrinsic functions, active culling of healthy epithelial cells, enhanced EC growth, and even cytoplasmic shedding by infected ECs. This article reviews current knowledge of the molecular mechanisms involved in gut regeneration in several insect models (Drosophila and Aedes of the order Diptera, and several Lepidoptera).
Collapse
Affiliation(s)
- Peng Zhang
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| | - Bruce A Edgar
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
43
|
Sensing microbial infections in the Drosophila melanogaster genetic model organism. Immunogenetics 2022; 74:35-62. [DOI: 10.1007/s00251-021-01239-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022]
|
44
|
Eleftherianos I, Heryanto C, Bassal T, Zhang W, Tettamanti G, Mohamed A. Haemocyte-mediated immunity in insects: Cells, processes and associated components in the fight against pathogens and parasites. Immunology 2021; 164:401-432. [PMID: 34233014 PMCID: PMC8517599 DOI: 10.1111/imm.13390] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
The host defence of insects includes a combination of cellular and humoral responses. The cellular arm of the insect innate immune system includes mechanisms that are directly mediated by haemocytes (e.g., phagocytosis, nodulation and encapsulation). In addition, melanization accompanying coagulation, clot formation and wound healing, nodulation and encapsulation processes leads to the formation of cytotoxic redox-cycling melanin precursors and reactive oxygen and nitrogen species. However, demarcation between cellular and humoral immune reactions as two distinct categories is not straightforward. This is because many humoral factors affect haemocyte functions and haemocytes themselves are an important source of many humoral molecules. There is also a considerable overlap between cellular and humoral immune functions that span from recognition of foreign intruders to clot formation. Here, we review these immune reactions starting with the cellular mechanisms that limit haemolymph loss and participate in wound healing and clot formation and advancing to cellular functions that are critical in restricting pathogen movement and replication. This information is important because it highlights that insect cellular immunity is controlled by a multilayered system, different components of which are activated by different pathogens or during the different stages of the infection.
Collapse
Affiliation(s)
- Ioannis Eleftherianos
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Christa Heryanto
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Taha Bassal
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| | - Wei Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural BioengineeringKey Laboratory of Green Pesticide and Agricultural BioengineeringMinistry of EducationGuizhou UniversityGuiyangChina
| | - Gianluca Tettamanti
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
- BAT Center‐Interuniversity Center for Studies on Bioinspired Agro‐Environmental TechnologyUniversity of Napoli Federico IINapoliItaly
| | - Amr Mohamed
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| |
Collapse
|
45
|
Davila K, Setlur S, Doermann D, Kota BU, Govindaraju V. Chart Mining: A Survey of Methods for Automated Chart Analysis. IEEE TRANSACTIONS ON PATTERN ANALYSIS AND MACHINE INTELLIGENCE 2021; 43:3799-3819. [PMID: 32365018 DOI: 10.1109/tpami.2020.2992028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Charts are useful communication tools for the presentation of data in a visually appealing format that facilitates comprehension. There have been many studies dedicated to chart mining, which refers to the process of automatic detection, extraction and analysis of charts to reproduce the tabular data that was originally used to create them. By allowing access to data which might not be available in other formats, chart mining facilitates the creation of many downstream applications. This paper presents a comprehensive survey of approaches across all components of the automated chart mining pipeline, such as (i) automated extraction of charts from documents; (ii) processing of multi-panel charts; (iii) automatic image classifiers to collect chart images at scale; (iv) automated extraction of data from each chart image, for popular chart types as well as selected specialized classes; (v) applications of chart mining; and (vi) datasets for training and evaluation, and the methods that were used to build them. Finally, we summarize the main trends found in the literature and provide pointers to areas for further research in chart mining.
Collapse
|
46
|
Kramer J, Neves J, Koniikusic M, Jasper H, Lamba DA. Dpp/TGFβ-superfamily play a dual conserved role in mediating the damage response in the retina. PLoS One 2021; 16:e0258872. [PMID: 34699550 PMCID: PMC8547621 DOI: 10.1371/journal.pone.0258872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022] Open
Abstract
Retinal homeostasis relies on intricate coordination of cell death and survival in response to stress and damage. Signaling mechanisms that coordinate this process in the adult retina remain poorly understood. Here we identify Decapentaplegic (Dpp) signaling in Drosophila and its mammalian homologue Transforming Growth Factor-beta (TGFβ) superfamily, that includes TGFβ and Bone Morphogenetic Protein (BMP) signaling arms, as central mediators of retinal neuronal death and tissue survival following acute damage. Using a Drosophila model for UV-induced retinal damage, we show that Dpp released from immune cells promotes tissue loss after UV-induced retinal damage. Interestingly, we find a dynamic response of retinal cells to this signal: in an early phase, Dpp-mediated stimulation of Saxophone/Smox signaling promotes apoptosis, while at a later stage, stimulation of the Thickveins/Mad axis promotes tissue repair and survival. This dual role is conserved in the mammalian retina through the TGFβ/BMP signaling, as supplementation of BMP4 or inhibition of TGFβ using small molecules promotes retinal cell survival, while inhibition of BMP negatively affects cell survival after light-induced photoreceptor damage and NMDA induced inner retinal neuronal damage. Our data identify key evolutionarily conserved mechanisms by which retinal homeostasis is maintained.
Collapse
Affiliation(s)
- Joshua Kramer
- Department of Ophthalmology, University of California, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, United States of America
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Joana Neves
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Faculdade de Medicina, Instituto de Medicina Molecular (iMM), Universidade de Lisboa, Lisbon, Portugal
| | - Mia Koniikusic
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Immunology Discovery, Genentech, Inc., South San Francisco, CA, United States of America
| | - Deepak A. Lamba
- Department of Ophthalmology, University of California, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, United States of America
- Buck Institute for Research on Aging, Novato, CA, United States of America
| |
Collapse
|
47
|
Kim J, Chuang HC, Wolf NK, Nicolai CJ, Raulet DH, Saijo K, Bilder D. Tumor-induced disruption of the blood-brain barrier promotes host death. Dev Cell 2021; 56:2712-2721.e4. [PMID: 34496290 DOI: 10.1016/j.devcel.2021.08.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
Cancer patients often die from symptoms that manifest at a distance from any tumor. Mechanisms underlying these systemic physiological perturbations, called paraneoplastic syndromes, may benefit from investigation in non-mammalian systems. Using a non-metastatic Drosophila adult model, we find that malignant-tumor-produced cytokines drive widespread host activation of JAK-STAT signaling and cause premature lethality. STAT activity is particularly high in cells of the blood-brain barrier (BBB), where it induces aberrant BBB permeability. Remarkably, inhibiting STAT in the BBB not only rescues barrier function but also extends the lifespan of tumor-bearing hosts. We identify BBB damage in other pathological conditions that cause elevated inflammatory signaling, including obesity and infection, where BBB permeability also regulates host survival. IL-6-dependent BBB dysfunction is further seen in a mouse tumor model, and it again promotes host morbidity. Therefore, BBB alterations constitute a conserved lethal tumor-host interaction that also underlies other physiological morbidities.
Collapse
Affiliation(s)
- Jung Kim
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Hsiu-Chun Chuang
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Natalie K Wolf
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Christopher J Nicolai
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - David H Raulet
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
48
|
Wang J, Liu Q, Gong Y, Jin LH. Anchor maintains gut homeostasis by restricting the JNK and Notch pathways in Drosophila. JOURNAL OF INSECT PHYSIOLOGY 2021; 134:104309. [PMID: 34496279 DOI: 10.1016/j.jinsphys.2021.104309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
The adult Drosophila intestinal epithelium must be tightly regulated to maintain regeneration and homeostasis. The dysregulation of the regenerative capacity is frequently associated with intestinal diseases such as inflammation and tumorigenesis. Here, we showed that the G protein-coupled receptor Anchor maintains Drosophila adult midgut homeostasis by restricting Jun-N-terminal kinase (JNK) and Notch pathway activity. anchor inactivation resulted in aberrant JNK pathway activation, which led to excessive enteroblast (EB) production and premature enterocyte (EC) differentiation. In addition, increased Notch levels promoted premature EC differentiation following the loss of anchor. This defect induced by the loss of anchor ultimately caused sensitivity to stress or environmental challenge in adult flies. Taken together, our results demonstrate that the activity of anchor is essential to coordinate stem cell differentiation and proliferation to maintain intestinal homeostasis.
Collapse
Affiliation(s)
- Jiewei Wang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Qiang Liu
- Department of Cell Biology and Genetics, Shenyang Medical College, 146 Huanghe North Street, Shenyang 110034, China
| | - Yuan Gong
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, No. 26 Hexing Road, Xiangfang District, Harbin 150040, China.
| |
Collapse
|
49
|
Cai XT, Li H, Jensen MB, Maksoud E, Borneo J, Liang Y, Quake SR, Luo L, Haghighi P, Jasper H. Gut cytokines modulate olfaction through metabolic reprogramming of glia. Nature 2021; 596:97-102. [PMID: 34290404 PMCID: PMC8911385 DOI: 10.1038/s41586-021-03756-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
Infection-induced aversion against enteropathogens is a conserved sickness behaviour that can promote host survival1,2. The aetiology of this behaviour remains poorly understood, but studies in Drosophila have linked olfactory and gustatory perception to avoidance behaviours against toxic microorganisms3-5. Whether and how enteric infections directly influence sensory perception to induce or modulate such behaviours remains unknown. Here we show that enteropathogen infection in Drosophila can modulate olfaction through metabolic reprogramming of ensheathing glia of the antennal lobe. Infection-induced unpaired cytokine expression in the intestine activates JAK-STAT signalling in ensheathing glia, inducing the expression of glial monocarboxylate transporters and the apolipoprotein glial lazarillo (GLaz), and affecting metabolic coupling of glia and neurons at the antennal lobe. This modulates olfactory discrimination, promotes the avoidance of bacteria-laced food and increases fly survival. Although transient in young flies, gut-induced metabolic reprogramming of ensheathing glia becomes constitutive in old flies owing to age-related intestinal inflammation, which contributes to an age-related decline in olfactory discrimination. Our findings identify adaptive glial metabolic reprogramming by gut-derived cytokines as a mechanism that causes lasting changes in a sensory system in ageing flies.
Collapse
Affiliation(s)
- Xiaoyu Tracy Cai
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.,Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,University of Southern California, Los Angeles, CA 90007, USA
| | - Hongjie Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA,Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Martin Borch Jensen
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,Gordian Biotechnology, 953 Indiana St., San Francisco, CA 94107, USA
| | - Elie Maksoud
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Jovencio Borneo
- FACS lab, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yuxin Liang
- NGS lab, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stephen R. Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA,Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Pejmun Haghighi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.,Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,Corresponding author: Heinrich Jasper, Genentech, 1 DNA Way, South San Francisco, California 94080, USA.
| |
Collapse
|
50
|
Leite THJF, Ferreira ÁGA, Imler JL, Marques JT. Distinct Roles of Hemocytes at Different Stages of Infection by Dengue and Zika Viruses in Aedes aegypti Mosquitoes. Front Immunol 2021; 12:660873. [PMID: 34093550 PMCID: PMC8169962 DOI: 10.3389/fimmu.2021.660873] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
Aedes aegypti mosquitoes are vectors for arboviruses of medical importance such as dengue (DENV) and Zika (ZIKV) viruses. Different innate immune pathways contribute to the control of arboviruses in the mosquito vector including RNA interference, Toll and Jak-STAT pathways. However, the role of cellular responses mediated by circulating macrophage-like cells known as hemocytes remains unclear. Here we show that hemocytes are recruited to the midgut of Ae. aegypti mosquitoes in response to DENV or ZIKV. Blockade of the phagocytic function of hemocytes using latex beads induced increased accumulation of hemocytes in the midgut and a reduction in virus infection levels in this organ. In contrast, inhibition of phagocytosis by hemocytes led to increased systemic dissemination and replication of DENV and ZIKV. Hence, our work reveals a dual role for hemocytes in Ae. aegypti mosquitoes, whereby phagocytosis is not required to control viral infection in the midgut but is essential to restrict systemic dissemination. Further understanding of the mechanism behind this duality could help the design of vector-based strategies to prevent transmission of arboviruses.
Collapse
Affiliation(s)
- Thiago H J F Leite
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Álvaro G A Ferreira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Brazil
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| | - João T Marques
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| |
Collapse
|