1
|
Ashaolu OP, Odukanmi OA, Salami AT, Abolaji AO, Olaleye SB. Kolaviron: a Bioflavonoid from Seed Extract of Garcinia kola Attenuates Chromium (VI)-Induced Gut Dysfunction and Oxidative Damage in Drosophila melanogaster. Biol Trace Elem Res 2024:10.1007/s12011-024-04414-y. [PMID: 39425879 DOI: 10.1007/s12011-024-04414-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Hexavalent chromium [Cr(VI)] is of public health significance due to its toxicity and carcinogenic effects. Kolaviron, a bio-flavonoid fraction from Garcinia kola seed, has been reported to possess gastroprotective and antioxidative properties. We hypothesize that Kolaviron-fortified diet will attenuate chromium (VI)-induced gut dysfunction and oxidative damage in Drosophila melanogaster. We exposed D. melanogaster (Oregon strain of 1-3 days old of both male and female) to a 1.0 mg/kg diet of chromium (VI), with or without Kolaviron (100 mg/kg diet) orally for 5 days. We evaluated markers of oxidative stress (total peroxide and protein carbonyl), antioxidative status (total thiols (T-SH), non-protein thiols (NP-SH), and catalase), and inflammatory (nitric oxide (nitrite and nitrate) and gut's morphology. The data indicated that Kolaviron ameliorated chromium (VI)-induced reduction in the levels of T-SH, NP-SH, and catalase activity (p < 0.05). In addition, Kolaviron attenuated chromium (VI)-induced elevation of total peroxide, protein carbonyl, and nitric oxide (p < 0.05). Kolaviron offered a protective role in chromium VI-induced toxicity in the gut of D. melanogaster. This study provided further insights into the protective mechanisms of Kolaviron against chromium (VI)-induced toxicity in D. melanogaster by maintaining epithelial integrity of the gut and oxidative stress-antioxidant balance.
Collapse
Affiliation(s)
- Onaara Peter Ashaolu
- Gastrointestinal Secretion and Inflammation Research Unit, Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Drosophila Research and Training Centre, Ibadan, Nigeria
| | - Olugbenga Adeola Odukanmi
- Gastrointestinal Secretion and Inflammation Research Unit, Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeola Temitope Salami
- Gastrointestinal Secretion and Inflammation Research Unit, Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Amos Olalekan Abolaji
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
- Drosophila Research and Training Centre, Ibadan, Nigeria.
| | - Samuel Babafemi Olaleye
- Gastrointestinal Secretion and Inflammation Research Unit, Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
2
|
Lin YC, Wu T, Wu CL. The Neural Correlations of Olfactory Associative Reward Memories in Drosophila. Cells 2024; 13:1716. [PMID: 39451234 PMCID: PMC11506542 DOI: 10.3390/cells13201716] [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: 09/20/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Advancing treatment to resolve human cognitive disorders requires a comprehensive understanding of the molecular signaling pathways underlying learning and memory. While most organ systems evolved to maintain homeostasis, the brain developed the capacity to perceive and adapt to environmental stimuli through the continuous modification of interactions within a gene network functioning within a broader neural network. This distinctive characteristic enables significant neural plasticity, but complicates experimental investigations. A thorough examination of the mechanisms underlying behavioral plasticity must integrate multiple levels of biological organization, encompassing genetic pathways within individual neurons, interactions among neural networks providing feedback on gene expression, and observable phenotypic behaviors. Model organisms, such as Drosophila melanogaster, which possess more simple and manipulable nervous systems and genomes than mammals, facilitate such investigations. The evolutionary conservation of behavioral phenotypes and the associated genetics and neural systems indicates that insights gained from flies are pertinent to understanding human cognition. Rather than providing a comprehensive review of the entire field of Drosophila memory research, we focus on olfactory associative reward memories and their related neural circuitry in fly brains, with the objective of elucidating the underlying neural mechanisms, thereby advancing our understanding of brain mechanisms linked to cognitive systems.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Tony Wu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City 23652, Taiwan;
| | - Chia-Lin Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City 23652, Taiwan;
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
3
|
Ignatiou A, Pitsouli C. Host-diet-microbiota interplay in intestinal nutrition and health. FEBS Lett 2024; 598:2482-2517. [PMID: 38946050 DOI: 10.1002/1873-3468.14966] [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: 04/21/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
The intestine is populated by a complex and dynamic assortment of microbes, collectively called gut microbiota, that interact with the host and contribute to its metabolism and physiology. Diet is considered a key regulator of intestinal microbiota, as ingested nutrients interact with and shape the resident microbiota composition. Furthermore, recent studies underscore the interplay of dietary and microbiota-derived nutrients, which directly impinge on intestinal stem cells regulating their turnover to ensure a healthy gut barrier. Although advanced sequencing methodologies have allowed the characterization of the human gut microbiome, mechanistic studies assessing diet-microbiota-host interactions depend on the use of genetically tractable models, such as Drosophila melanogaster. In this review, we first discuss the similarities between the human and fly intestines and then we focus on the effects of diet and microbiota on nutrient-sensing signaling cascades controlling intestinal stem cell self-renewal and differentiation, as well as disease. Finally, we underline the use of the Drosophila model in assessing the role of microbiota in gut-related pathologies and in understanding the mechanisms that mediate different whole-body manifestations of gut dysfunction.
Collapse
Affiliation(s)
- Anastasia Ignatiou
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
4
|
Christou M, Fidelix A, Apidianakis Y, Andreou C. Nanoparticle Uptake in the Aging and Oncogenic Drosophila Midgut Measured with Surface-Enhanced Raman Spectroscopy. Cells 2024; 13:1344. [PMID: 39195234 DOI: 10.3390/cells13161344] [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: 06/11/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024] Open
Abstract
Colorectal cancer remains a major global health concern. Colonoscopy, the gold-standard colorectal cancer diagnostic, relies on the visual detection of lesions and necessitates invasive biopsies for confirmation. Alternative diagnostic methods, based on nanomedicine, can facilitate early detection of malignancies. Here, we examine the uptake of surface-enhanced Raman scattering nanoparticles (SERS NPs) as a marker for intestinal tumor detection and imaging using an established Drosophila melanogaster model for gut disease. Young and old Oregon-R and w1118 flies were orally administered SERS NPs and scanned without and upon gut lumen clearance to assess nanoparticle retention as a function of aging. Neither young nor old flies showed significant NP retention in their body after gut lumen clearance. Moreover, tumorigenic flies of the esg-Gal4/UAS-RasV12 genotype were tested for SERS NP retention 2, 4 and 6 days after RasV12 oncogene induction in their midgut progenitor cells. Tumorigenic flies showed a statistically significant NP retention signal at 2 days, well before midgut epithelium impairment. The signal was then visualized in scans of dissected guts revealing areas of NP uptake in the posterior midgut region of high stem cell activity.
Collapse
Affiliation(s)
- Maria Christou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus
| | - Ayobami Fidelix
- Department of Electrical and Computer Engineering, University of Cyprus, Nicosia 2109, Cyprus
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus
| | - Chrysafis Andreou
- Department of Electrical and Computer Engineering, University of Cyprus, Nicosia 2109, Cyprus
| |
Collapse
|
5
|
Pratama MR, Wahyudin E, Putri TZ, Hardiyanti W, Fatiah D, Chaeratunnisa R, Bapulo NN, Latada NP, Mudjahid M, Nainu F. A fruit fly-based approach to unraveling enteropathy-causing pharmaceuticals. NARRA J 2024; 4:e898. [PMID: 39280279 PMCID: PMC11394174 DOI: 10.52225/narra.v4i2.898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Enteropathy is a gastrointestinal disorder characterized by inflammation in the small intestine and one of the causes of enteropathy is the side effects of certain drugs, such as non-steroidal anti-inflammatory drugs (NSAIDs). The mechanism of NSAIDs, such as indomethacin, could inhibit prostaglandin synthesis, leading to a decrease in mucus production and small intestine integrity. To test the effects of a drug, it is necessary to undergo preclinical testing using animal models. Commonly used animal models such as mice and rats have several drawbacks including high cost, ethical issues, and long lifespan. Therefore, alternatives such as using invertebrate animals like Drosophila melanogaster as a more economical in vivo platform with genetic similarity to mammals and devoid of ethical concerns are needed. The aim of this study was to evaluate Drosophila melanogaster as an in vivo model organism in testing the side effects of pharmaceuticals that cause enteropathy. In this study, flies aged 3-5 days were starved and then placed into treatment vials comprising untreated control and indomethacin-treated (3.75 mM, 7.5 mM, and 15 mM). Survival analysis was conducted during the treatment period, followed by a Smurf assay test after seven days of treatment. Subsequently, the expression of pro-inflammatory cytokine-related genes (drs and totA), mitochondria stability-related genes (tom40), and endogenous antioxidant-related genes (sod1, sod2, and cat) was performed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Our data indicated that indomethacin did not impact lifespan or cause intestinal damage. However, we observed increased expression of pro-inflammatory cytokine-related genes, including drs, and a twofold increase in totA gene expression. Furthermore, there was a significant upregulation of mitochondrial stability gene tom40, endogenous antioxidant genes sod1 and cat, and a threefold increase in sod2 at 15 mM indomethacin. Although no phenotypical changes in gut integrity were detected, the increased expression of pro-inflammatory cytokine genes suggests the occurrence of inflammation in the indomethacin-treated flies.
Collapse
Affiliation(s)
- Muhammad R Pratama
- Postgraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Elly Wahyudin
- Departmentof Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Tenri Zad Putri
- Postgraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Widya Hardiyanti
- Postgraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Dewita Fatiah
- Postgraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Rizkya Chaeratunnisa
- Undergraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Nurdewi N Bapulo
- Postgraduate Program in Biomedical Science, Graduate School of Medicine, Universitas Hasanuddin, Makassar, Indonesia
| | - Nadila P Latada
- Unhas Fly Research Group, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Mukarram Mudjahid
- Departmentof Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Firzan Nainu
- Departmentof Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
- Unhas Fly Research Group, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| |
Collapse
|
6
|
Sadova N, Blank-Landeshammer B, Curic D, Iken M, Weghuber J. Sex-specific pharmacokinetic response to phytoestrogens in Drosophila melanogaster. Biomed Pharmacother 2024; 175:116612. [PMID: 38663102 DOI: 10.1016/j.biopha.2024.116612] [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: 02/13/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 06/03/2024] Open
Abstract
Drosophila melanogaster, or the fruit fly, is widely used for modeling numerous human diseases, such as neurodegeneration, tumor development, cachexia, and intestinal dysfunction. It is a suitable model organism for research targeting the physiology and pathophysiology of the intestinal epithelial barrier and has also been used as a model organism for preliminary drug and bioactive nutrient screening. However, the application of D. melanogaster in research on drug bioavailability and pharmacokinetic properties has not yet been well explored. In this study, we applied D. melanogaster to investigate the absorption and excretion of the orally administered phytoestrogens daidzein, glycitein, genistein, and their glycosides. Therefore, we established a quick, noninvasive method to quantify compound retention in D. melanogaster, suitable for the investigation of a broad variety of potentially bioactive substances. We showed that fruit fly sex plays a key role in the metabolization, transportation, and excretion of phytoestrogenic isoflavones. In particular, female fruit flies retained significantly more isoflavones than male fruit flies, which was reflected in the greater metabolic impact of isoflavones on females. Male fruit flies excreted more isoflavones than females did, which was linked to the upregulation of the xenobiotic transporter gene Mdr50. We also demonstrated that micellized isoflavones were more bioavailable than powdered isoflavones, independent of sex, age or the addition of dietary fibers.
Collapse
Affiliation(s)
- Nadiia Sadova
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels 4600, Austria
| | - Bernhard Blank-Landeshammer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels 4600, Austria; FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln 3430, Austria
| | - David Curic
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels 4600, Austria
| | - Marcus Iken
- PM International AG, Schengen, Luxembourg 5445, Luxembourg
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels 4600, Austria; FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln 3430, Austria.
| |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 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
|
8
|
Alaraby M, Abass D, Farre M, Hernández A, Marcos R. Are bioplastics safe? Hazardous effects of polylactic acid (PLA) nanoplastics in Drosophila. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170592. [PMID: 38354814 DOI: 10.1016/j.scitotenv.2024.170592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
The expanded uses of bioplastics require understanding the potential health risks associated with their exposure. To address this issue, Drosophila melanogaster as a versatile terrestrial in vivo model was employed, and polylactic acid nanoplastics (PLA-NPLs), as a proxy for bioplastics, were tested as a material model. Effects were determined in larvae exposed for 4 days to different concentrations (25, 100, and 400 μg/mL) of 463.9 ± 129.4 nm PLA-NPLs. Transmission electron microscopy (TEM) and scanning electron microscope (SEM) approaches permitted the detection of PLA-NPLs in the midgut lumen of Drosophila larvae, interacting with symbiotic bacteria. Enzymatic vacuoles were observed as carriers, collecting PLA-NPLs and enabling the crossing of the peritrophic membrane, finally internalizing into enterocytes. Although no toxic effects were observed in egg-to-adult survival, cell uptake of PLA-NPLs causes cytological disturbances and the formation of large vacuoles. The translocation across the intestinal barrier was demonstrated by their presence in the hemolymph. PLA-NPL exposure triggered intestinal damage, oxidative stress, DNA damage, and inflammation responses, as evaluated via a wide set of marker genes. Collectively, these structural and molecular interferences caused by PLA-NPLs generated high levels of oxidative stress and DNA damage in the hemocytes of Drosophila larvae. The observed effects point out the need for further studies aiming to deepen the health risks of bioplastics before adopting their uses as a safe plastic alternative.
Collapse
Affiliation(s)
- Mohamed Alaraby
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Zoology Department, Faculty of Sciences, Sohag University, 82524 Sohag, Egypt.
| | - Doaa Abass
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Zoology Department, Faculty of Sciences, Sohag University, 82524 Sohag, Egypt
| | - Marinella Farre
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDÆA-CSIC), 08034 Barcelona, Spain
| | - Alba Hernández
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ricard Marcos
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| |
Collapse
|
9
|
Miles J, Lozano GL, Rajendhran J, Stabb EV, Handelsman J, Broderick NA. Massively parallel mutant selection identifies genetic determinants of Pseudomonas aeruginosa colonization of Drosophila melanogaster. mSystems 2024; 9:e0131723. [PMID: 38380971 PMCID: PMC10949475 DOI: 10.1128/msystems.01317-23] [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: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 02/22/2024] Open
Abstract
Pseudomonas aeruginosa is recognized for its ability to colonize diverse habitats and cause disease in a variety of hosts, including plants, invertebrates, and mammals. Understanding how this bacterium is able to occupy wide-ranging niches is important for deciphering its ecology. We used transposon sequencing [Tn-Seq, also known as insertion sequencing (INSeq)] to identify genes in P. aeruginosa that contribute to fitness during the colonization of Drosophila melanogaster. Our results reveal a suite of critical factors, including those that contribute to polysaccharide production, DNA repair, metabolism, and respiration. Comparison of candidate genes with fitness determinants discovered in previous studies on P. aeruginosa identified several genes required for colonization and virulence determinants that are conserved across hosts and tissues. This analysis provides evidence for both the conservation of function of several genes across systems, as well as host-specific functions. These findings, which represent the first use of transposon sequencing of a gut pathogen in Drosophila, demonstrate the power of Tn-Seq in the fly model system and advance the existing knowledge of intestinal pathogenesis by D. melanogaster, revealing bacterial colonization determinants that contribute to a comprehensive portrait of P. aeruginosa lifestyles across habitats.IMPORTANCEDrosophila melanogaster is a powerful model for understanding host-pathogen interactions. Research with this system has yielded notable insights into mechanisms of host immunity and defense, many of which emerged from the analysis of bacterial mutants defective for well-characterized virulence factors. These foundational studies-and advances in high-throughput sequencing of transposon mutants-support unbiased screens of bacterial mutants in the fly. To investigate mechanisms of host-pathogen interplay and exploit the tractability of this model host, we used a high-throughput, genome-wide mutant analysis to find genes that enable the pathogen P. aeruginosa to colonize the fly. Our analysis reveals critical mediators of P. aeruginosa establishment in its host, some of which are required across fly and mouse systems. These findings demonstrate the utility of massively parallel mutant analysis and provide a platform for aligning the fly toolkit with comprehensive bacterial genomics.
Collapse
Affiliation(s)
- Jessica Miles
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
- Graduate Program in Microbiology, Yale University, New Haven, Connecticut, USA
| | - Gabriel L. Lozano
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Jeyaprakash Rajendhran
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Eric V. Stabb
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois, USA
| | - Jo Handelsman
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|
10
|
Umargamwala R, Manning J, Dorstyn L, Denton D, Kumar S. Understanding Developmental Cell Death Using Drosophila as a Model System. Cells 2024; 13:347. [PMID: 38391960 PMCID: PMC10886741 DOI: 10.3390/cells13040347] [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: 01/23/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Cell death plays an essential function in organismal development, wellbeing, and ageing. Many types of cell deaths have been described in the past 30 years. Among these, apoptosis remains the most conserved type of cell death in metazoans and the most common mechanism for deleting unwanted cells. Other types of cell deaths that often play roles in specific contexts or upon pathological insults can be classed under variant forms of cell death and programmed necrosis. Studies in Drosophila have contributed significantly to the understanding and regulation of apoptosis pathways. In addition to this, Drosophila has also served as an essential model to study the genetic basis of autophagy-dependent cell death (ADCD) and other relatively rare types of context-dependent cell deaths. Here, we summarise what is known about apoptosis, ADCD, and other context-specific variant cell death pathways in Drosophila, with a focus on developmental cell death.
Collapse
Affiliation(s)
- Ruchi Umargamwala
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Jantina Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Loretta Dorstyn
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
11
|
Huang WH, Kajal K, Wibowo RH, Amartuvshin O, Kao SH, Rastegari E, Lin CH, Chiou KL, Pi HW, Ting CT, Hsu HJ. Excess dietary sugar impairs Drosophila adult stem cells via elevated reactive oxygen species-induced JNK signaling. Development 2024; 151:dev201772. [PMID: 38063853 DOI: 10.1242/dev.201772] [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: 03/16/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
High-sugar diets (HSDs) often lead to obesity and type 2 diabetes, both metabolic syndromes associated with stem cell dysfunction. However, it is unclear whether excess dietary sugar affects stem cells. Here, we report that HSD impairs stem cell function in the intestine and ovaries of female Drosophila prior to the onset of insulin resistance, a hallmark of type 2 diabetes. Although 1 week of HSD leads to obesity, impaired oogenesis and altered lipid metabolism, insulin resistance does not occur. HSD increases glucose uptake by germline stem cells (GSCs) and triggers reactive oxygen species-induced JNK signaling, which reduces GSC proliferation. Removal of excess sugar from the diet reverses these HSD-induced phenomena. A similar phenomenon is found in intestinal stem cells (ISCs), except that HSD disrupts ISC maintenance and differentiation. Interestingly, tumor-like GSCs and ISCs are less responsive to HSD, which may be because of their dependence on glycolytic metabolism and high energy demand, respectively. This study suggests that excess dietary sugar induces oxidative stress and damages stem cells before insulin resistance develops, a mechanism that may also occur in higher organisms.
Collapse
Affiliation(s)
- Wei-Hao Huang
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Department of Life Science, National Taiwan University, Taipei 10917
| | - Kreeti Kajal
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei 11529
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227
| | | | - Oyundari Amartuvshin
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei 11529
- Graduate Institute of Life Science, National Defense Medical Center, Taipei 11490
| | - Shih-Han Kao
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| | - Elham Rastegari
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| | - Chi-Hung Lin
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei 11529
- Graduate Institute of Life Science, National Defense Medical Center, Taipei 11490
| | - Kuan-Lin Chiou
- Department of Biomedical Science, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Hai-Wei Pi
- Department of Biomedical Science, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Chau-Ti Ting
- Department of Life Science, National Taiwan University, Taipei 10917
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| |
Collapse
|
12
|
Usman D, Abubakar MB, Ibrahim KG, Imam MU. Iron chelation and supplementation: A comparison in the management of inflammatory bowel disease using drosophila. Life Sci 2024; 336:122328. [PMID: 38061132 DOI: 10.1016/j.lfs.2023.122328] [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: 09/23/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
AIMS Inflammatory Bowel Disease (IBD) is associated with systemic iron deficiency and has been managed with iron supplements which cause adverse side effects. Conversely, some reports highlight iron depletion to ameliorate IBD. The underlying intestinal response and comparative benefit of iron depletion and supplementation in IBD is unknown. The aims of this work were to characterize and compare the effects of iron supplementation and iron depletion in IBD. MAIN METHODS IBD was induced in Drosophila melanogaster using 3 % dextran sodium sulfate (DSS) in diet for 7 days. Using this model, we investigated the impacts of acute iron depletion (using bathophenanthroline disulfonate, BPS) and supplementation (using ferrous sulphate, FS), before and after IBD induction, on gut iron homeostasis, cell death, gut permeability, inflammation, antioxidant defence, antimicrobial response and several fly phenotypes. KEY FINDINGS DSS decreased fly mass (p < 0.001), increased gut permeability (p < 0.001) and shortened lifespan (p = 0.035) compared to control. The DSS-fed flies also showed significantly elevated lipid peroxidation (p < 0.001), and the upregulated expression of apoptotic marker- drice (p < 0.001), tight junction protein - bbg (p < 0.001), antimicrobial peptide - dpta (p = 0.002) and proinflammatory cytokine - upd2 (p < 0.001). BPS significantly (p < 0.05) increased fly mass and lifespan, decreased gut permeability, decreased lipid peroxidation and decreased levels of drice, bbg, dpta and upd2 in IBD flies. This iron chelation (using BPS) showed better protection from DSS-induced IBD than iron supplementation (using FS). Preventive and curative interventions, by BPS or FS, also differed in outcomes. SIGNIFICANCE This may inform precise management strategies aimed at tackling IBD and its recurrence.
Collapse
Affiliation(s)
- Dawoud Usman
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Physiology, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Murtala Bello Abubakar
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Physiology, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Baze University, Abuja, Nigeria
| | - Kasimu Ghandi Ibrahim
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Physiology, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, P. O. Box 2000, Zarqa 13110, Jordan; School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa
| | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria; Department of Medical Biochemistry, College of Health Sciences, Usmanu Danfodiyo University Sokoto, Nigeria.
| |
Collapse
|
13
|
Wang S, Li Y, Lin X, Fu X, Zhong H, Ren K, Liu C, Yao W. Rapid Screening of Phenolic Compounds with Anti-Enteritis Activity from Camellia oleifera Oil Using a Smurf Drosophila Model and Molecular Docking Methods. Molecules 2023; 29:76. [PMID: 38202658 PMCID: PMC10780214 DOI: 10.3390/molecules29010076] [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: 10/31/2023] [Revised: 12/03/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Screening and identifying the active compounds in foods are important for the development and utilization of functional foods. In this study, the anti-enteritis activity of ethanol extract from Camellia oleifera oil (PECS) was quickly evaluated using a Smurf Drosophila model and the metabolomics approach, combined with molecular docking techniques, were performed to rapidly screen and identify compounds with potential anti-enteritis activity in PECS. PECS showed good anti-enteritis activity and inhibited the activity of 5-lipoxygenase (LOX), cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS). In particular, wighteone and p-octopamine were newly identified in C. oleifera oil and were proven to have good anti-enteritis activity. The inhibitory activity of kaempferitrin (IC50 = 0.365 mmol L-1) was higher than that of wighteone (IC50 = 0.424 mmol L-1) and p-octopamine (IC50 = 0.402 mmol L-1). Of note, the IC50 value of salazosulfapyridine was 0.810 mmol L-1. Inhibition of LOX activity is likely one of the anti-enteritis mechanisms of PECS. These new findings lay the foundation for further investigations into the underlying mechanisms of anti-enteritis activity in C. oleifera oil.
Collapse
Affiliation(s)
- Shuhao Wang
- Hunan Provincial Key Laboratory of Forest Edible Resources Safety and Processing Utilization, Central South University of Forestry and Technology, Changsha 410004, China
- Commodity Quality Inspection Institute of Hunan Province, Changsha 410004, China
| | - Yang Li
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Xin Lin
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Xiangjin Fu
- Hunan Provincial Key Laboratory of Forest Edible Resources Safety and Processing Utilization, Central South University of Forestry and Technology, Changsha 410004, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Haiyan Zhong
- Hunan Provincial Key Laboratory of Forest Edible Resources Safety and Processing Utilization, Central South University of Forestry and Technology, Changsha 410004, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Kangzi Ren
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Cheng Liu
- Yi-Feng Agriculture and Forestry Technology Co., Ltd., Shaoyang 422300, China
| | - Wen Yao
- Hunan Ju Xiong Institute of Camellia oleifera Oil, Yueyang 414000, China;
| |
Collapse
|
14
|
Miles J, Lozano GL, Rajendhran J, Stabb EV, Handelsman J, Broderick NA. Massively parallel mutant selection identifies genetic determinants of Pseudomonas aeruginosa colonization of Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567573. [PMID: 38045230 PMCID: PMC10690197 DOI: 10.1101/2023.11.20.567573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Pseudomonas aeruginosa is recognized for its ability to colonize diverse habitats and cause disease in a variety of hosts, including plants, invertebrates, and mammals. Understanding how this bacterium is able to occupy wide-ranging niches is important for deciphering its ecology. We used transposon sequencing (Tn-Seq, also known as INSeq) to identify genes in P. aeruginosa that contribute to fitness during colonization of Drosophila melanogaster. Our results reveal a suite of critical factors, including those that contribute to polysaccharide production, DNA repair, metabolism, and respiration. Comparison of candidate genes with fitness determinants discovered in previous studies of P. aeruginosa identified several genes required for colonization and virulence determinants that are conserved across hosts and tissues. This analysis provides evidence for both the conservation of function of several genes across systems, as well as host-specific functions. These findings, which represent the first use of transposon sequencing of a gut pathogen in Drosophila, demonstrate the power of Tn-Seq in the fly model system and advance existing knowledge of intestinal pathogenesis by D. melanogaster, revealing bacterial colonization determinants that contribute to a comprehensive portrait of P. aeruginosa lifestyles across habitats.
Collapse
Affiliation(s)
- Jessica Miles
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Graduate Program in Microbiology, Yale University, New Haven, CT, USA
| | - Gabriel L. Lozano
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Current address: Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeyaprakash Rajendhran
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Current address: Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, TN, India
| | - Eric V. Stabb
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Jo Handelsman
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
- Current address: Wisconsin Institute for Discovery and Department of Plant Pathology, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
15
|
Yun HM, Hyun S. Role of gut commensal bacteria in juvenile developmental growth of the host: insights from Drosophila studies. Anim Cells Syst (Seoul) 2023; 27:329-339. [PMID: 38023592 PMCID: PMC10653766 DOI: 10.1080/19768354.2023.2282726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
The gut microbiome plays a crucial role in maintaining health in a variety of organisms, from insects to humans. Further, beneficial symbiotic microbes are believed to contribute to improving the quality of life of the host. Drosophila is an optimal model for studying host-commensal microbe interactions because it allows for convenient manipulation of intestinal microbial composition. Fly microbiota has a simple taxonomic composition and can be cultivated and genetically tracked. This permits functional studies and analyses of the molecular mechanisms underlying their effects on host physiological processes. In this context, we briefly introduce the principle of juvenile developmental growth in Drosophila. Then, we discuss the current understanding of the molecular mechanisms underlying the effects of gut commensal bacteria, such as Lactiplantibacillus plantarum and Acetobacter pomorum, in the fly gut microbiome on Drosophila juvenile growth, including specific actions of gut hormones and metabolites in conserved cellular signaling systems, such as the insulin/insulin-like (IIS) and the target of rapamycin (TOR) pathways. Given the similarities in tissue function/structure, as well as the high conservation of physiological systems between Drosophila and mammals, findings from the Drosophila model system will have significant implications for understanding the mechanisms underlying the interaction between the host and the gut microbiome in metazoans.
Collapse
Affiliation(s)
- Hyun Myoung Yun
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| | - Seogang Hyun
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| |
Collapse
|
16
|
Cabasso O, Kuppuramalingam A, Lelieveld L, Van der Lienden M, Boot R, Aerts JM, Horowitz M. Animal Models for the Study of Gaucher Disease. Int J Mol Sci 2023; 24:16035. [PMID: 38003227 PMCID: PMC10671165 DOI: 10.3390/ijms242216035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
In Gaucher disease (GD), a relatively common sphingolipidosis, the mutant lysosomal enzyme acid β-glucocerebrosidase (GCase), encoded by the GBA1 gene, fails to properly hydrolyze the sphingolipid glucosylceramide (GlcCer) in lysosomes, particularly of tissue macrophages. As a result, GlcCer accumulates, which, to a certain extent, is converted to its deacylated form, glucosylsphingosine (GlcSph), by lysosomal acid ceramidase. The inability of mutant GCase to degrade GlcSph further promotes its accumulation. The amount of mutant GCase in lysosomes depends on the amount of mutant ER enzyme that shuttles to them. In the case of many mutant GCase forms, the enzyme is largely misfolded in the ER. Only a fraction correctly folds and is subsequently trafficked to the lysosomes, while the rest of the misfolded mutant GCase protein undergoes ER-associated degradation (ERAD). The retention of misfolded mutant GCase in the ER induces ER stress, which evokes a stress response known as the unfolded protein response (UPR). GD is remarkably heterogeneous in clinical manifestation, including the variant without CNS involvement (type 1), and acute and subacute neuronopathic variants (types 2 and 3). The present review discusses animal models developed to study the molecular and cellular mechanisms underlying GD.
Collapse
Affiliation(s)
- Or Cabasso
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel; (O.C.); (A.K.)
| | - Aparna Kuppuramalingam
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel; (O.C.); (A.K.)
| | - Lindsey Lelieveld
- Leiden Institute of Chemistry, Leiden University, 9502 Leiden, The Netherlands; (L.L.); (M.V.d.L.); (R.B.)
| | - Martijn Van der Lienden
- Leiden Institute of Chemistry, Leiden University, 9502 Leiden, The Netherlands; (L.L.); (M.V.d.L.); (R.B.)
| | - Rolf Boot
- Leiden Institute of Chemistry, Leiden University, 9502 Leiden, The Netherlands; (L.L.); (M.V.d.L.); (R.B.)
| | - Johannes M. Aerts
- Leiden Institute of Chemistry, Leiden University, 9502 Leiden, The Netherlands; (L.L.); (M.V.d.L.); (R.B.)
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel; (O.C.); (A.K.)
| |
Collapse
|
17
|
Hsieh TC, Chiang HC. IMD signaling in the gut and the brain modulates Amyloid-beta-induced deficits in Drosophila. Life Sci 2023; 332:122118. [PMID: 37741318 DOI: 10.1016/j.lfs.2023.122118] [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: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
AIMS Evidence indicates accumulating Aβ peptides in brain activates immune responses in neuronal and peripheral system, which may collaboratively influence pathogenesis of Alzheimer's disease (AD). We aim to investigate whether regulating intestinal innate immune signaling ameliorates Aβ-induced impairments in Drosophila melanogaster. MAIN METHODS Quantitative polymerase chain reaction (qPCR) was used to observe expression changes of innate immune responses related genes in brain and in gut under the circumstance of Aβ overexpressing in nerve system. Aversive olfactory conditioning and survival assay were used to investigate effects of modulating Attacin-A (AttA) and Dpitercin-A (DptA). Fluorometric assays of respiratory burst activity was introduced to explore whether reducing oxidative stress enables overexpressing intestinal AttA and DptA to reverse Aβ-induced deficits. KEY FINDINGS In vivo genetic analysis revealed that accumulating Aβ42 in neurons modulates innate immune signaling of the IMD pathway both in the brain and in the gut. Increased expression levels of the intestinal AttA and DptA improved learning performance and extended the lifespan of Aβ42 flies. The administration of apramycin led to alleviations of Aβ-induced behavioral changes, indicating that gram-negative bacteria are associated with the development of Aβ-induced pathologies. Further analysis showed that the neural expression of Aβ42 increased oxidative stress in the gut, which disrupted intestinal integrity and decreased learning performance. In addition, increased levels of AMPs targeting gram-negative bacteria and antioxidants reduced oxidative stress in the gut and reversed Aβ-induced behavioral damage. SIGNIFICANCE These findings suggest that innate immune responses in the gut play a pivotal role in modulating Aβ-induced pathologies.
Collapse
Affiliation(s)
- Tsung-Chi Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsueh-Cheng Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Keyan KS, Salim S, Gowda S, Abdelrahman D, Amir SS, Islam Z, Vargas C, Bengoechea-Alonso MT, Alwa A, Dahal S, Kolatkar PR, Da'as S, Torrisani J, Ericsson J, Mohammad F, Khan OM. Control of TGFβ signalling by ubiquitination independent function of E3 ubiquitin ligase TRIP12. Cell Death Dis 2023; 14:692. [PMID: 37863914 PMCID: PMC10589240 DOI: 10.1038/s41419-023-06215-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: 06/19/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
Transforming growth factor β (TGFβ) pathway is a master regulator of cell proliferation, differentiation, and death. Deregulation of TGFβ signalling is well established in several human diseases including autoimmune disorders and cancer. Thus, understanding molecular pathways governing TGFβ signalling may help better understand the underlying causes of some of those conditions. Here, we show that a HECT domain E3 ubiquitin ligase TRIP12 controls TGFβ signalling in multiple models. Interestingly, TRIP12 control of TGFβ signalling is completely independent of its E3 ubiquitin ligase activity. Instead, TRIP12 recruits SMURF2 to SMAD4, which is most likely responsible for inhibitory monoubiquitination of SMAD4, since SMAD4 monoubiquitination and its interaction with SMURF2 were dramatically downregulated in TRIP12-/- cells. Additionally, genetic inhibition of TRIP12 in human and murine cells leads to robust activation of TGFβ signalling which was rescued by re-introducing wildtype TRIP12 or a catalytically inactive C1959A mutant. Importantly, TRIP12 control of TGFβ signalling is evolutionary conserved. Indeed, genetic inhibition of Drosophila TRIP12 orthologue, ctrip, in gut leads to a reduced number of intestinal stem cells which was compensated by the increase in differentiated enteroendocrine cells. These effects were completely normalised in Drosophila strain where ctrip was co-inhibited together with Drosophila SMAD4 orthologue, Medea. Similarly, in murine 3D intestinal organoids, CRISPR/Cas9 mediated genetic targeting of Trip12 enhances TGFβ mediated proliferation arrest and cell death. Finally, CRISPR/Cas9 mediated genetic targeting of TRIP12 in MDA-MB-231 breast cancer cells enhances the TGFβ induced migratory capacity of these cells which was rescued to the wildtype level by re-introducing wildtype TRIP12. Our work establishes TRIP12 as an evolutionary conserved modulator of TGFβ signalling in health and disease.
Collapse
Affiliation(s)
- Kripa S Keyan
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Safa Salim
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Swetha Gowda
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Syeda Sakina Amir
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Zeyaul Islam
- Qatar Biomedical Research Institute, Doha, Qatar
| | - Claire Vargas
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | | | - Amira Alwa
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Subrat Dahal
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Sahar Da'as
- Department of Research, Sidra Medicine, Doha, Qatar
| | - Jerome Torrisani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Johan Ericsson
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Farhan Mohammad
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Omar M Khan
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
19
|
Aalto AL, Saadabadi A, Lindholm F, Kietz C, Himmelroos E, Marimuthu P, Salo-Ahen OMH, Eklund P, Meinander A. Stilbenoid compounds inhibit NF-κB-mediated inflammatory responses in the Drosophila intestine. Front Immunol 2023; 14:1253805. [PMID: 37809071 PMCID: PMC10556681 DOI: 10.3389/fimmu.2023.1253805] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Stilbenoid compounds have been described to have anti-inflammatory properties in animal models in vivo, and have been shown to inhibit Ca2+-influx through the transient receptor potential ankyrin 1 (TrpA1). Methods To study how stilbenoid compounds affect inflammatory signaling in vivo, we have utilized the fruit fly, Drosophila melanogaster, as a model system. To induce intestinal inflammation in the fly, we have fed flies with the intestinal irritant dextran sodium sulphate (DSS). Results We found that DSS induces severe changes in the bacteriome of the Drosophila intestine, and that this dysbiosis causes activation of the NF-κB transcription factor Relish. We have taken advantage of the DSS-model to study the anti-inflammatory properties of the stilbenoid compounds pinosylvin (PS) and pinosylvin monomethyl ether (PSMME). With the help of in vivo approaches, we have identified PS and PSMME to be transient receptor ankyrin 1 (TrpA1)-dependent antagonists of NF-κB-mediated intestinal immune responses in Drosophila. We have also computationally predicted the putative antagonist binding sites of these compounds at Drosophila TrpA1. Discussion Taken together, we show that the stilbenoids PS and PSMME have anti-inflammatory properties in vivo in the intestine and can be used to alleviate chemically induced intestinal inflammation in Drosophila.
Collapse
Affiliation(s)
- Anna L. Aalto
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| | - Atefeh Saadabadi
- Pharmaceutical Sciences Laboratory, Pharmacy, Åbo Akademi University, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, Turku, Finland
- Laboratory of Molecular Science and Engineering, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Fanny Lindholm
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Christa Kietz
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Emmy Himmelroos
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Parthiban Marimuthu
- Pharmaceutical Sciences Laboratory, Pharmacy, Åbo Akademi University, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, Turku, Finland
| | - Outi M. H. Salo-Ahen
- Pharmaceutical Sciences Laboratory, Pharmacy, Åbo Akademi University, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, Turku, Finland
| | - Patrik Eklund
- Laboratory of Molecular Science and Engineering, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Annika Meinander
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| |
Collapse
|
20
|
Qush A, Al Khatib HA, Rachid H, Al-Tamimi H, Al-Eshaq A, Al-Adwi S, Yassine HM, Kamareddine L. Intake of caffeine containing sugar diet remodels gut microbiota and perturbs Drosophila melanogaster immunity and lifespan. Microbes Infect 2023; 25:105149. [PMID: 37169244 DOI: 10.1016/j.micinf.2023.105149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
The diet-microbiome-immunity axis is one among the many arms that draw up the "we are what we intake" proclamation. As such, studies on the effect of food and beverage intake on the gut environment and microbiome and on modulating immunological responses and the host's susceptibility to pathogens are on the rise. A typical accompaniment in different sustenance we consume on daily basis is the trimethylxanthine alkaloid caffeine. Being a chief component in our regular aliment, a better understanding of the effect of caffeine containing food and beverages on our gut-microbiome-immunity axis and henceforth on our health is much needed. In this study, we shed more light on the effect of oral consumption of caffeine supplemented sugar diet on the gut environment, specifically on the gut microbiota, innate immunity and host susceptibility to pathogens using the Drosophila melanogaster model organism. Our findings reveal that the oral intake of a dose-specific caffeine containing sucrose/agarose sugar diet causes a significant alteration within the fly gut milieu demarcated by microbial dysbiosis and an elevation in the production of reactive oxygen species and expression of immune-deficiency (Imd) pathway-dependent antimicrobial peptide genes. The oral intake of caffeine containing sucrose/agarose sugar diet also renders the flies more susceptible to bacterial infection and shortens their lifespan in both infection and non-infection settings. Our findings set forth additional insight into the potentiality of diet to alter the gut milieu and highlight the importance of dietary control on health.
Collapse
Affiliation(s)
- Abeer Qush
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hebah A Al Khatib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Hajar Rachid
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hend Al-Tamimi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Alyaa Al-Eshaq
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Shaima Al-Adwi
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Layla Kamareddine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
21
|
Erazo-Oliveras A, Muñoz-Vega M, Mlih M, Thiriveedi V, Salinas ML, Rivera-Rodríguez JM, Kim E, Wright RC, Wang X, Landrock KK, Goldsby JS, Mullens DA, Roper J, Karpac J, Chapkin RS. Mutant APC reshapes Wnt signaling plasma membrane nanodomains by altering cholesterol levels via oncogenic β-catenin. Nat Commun 2023; 14:4342. [PMID: 37468468 PMCID: PMC10356786 DOI: 10.1038/s41467-023-39640-w] [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: 11/29/2021] [Accepted: 06/21/2023] [Indexed: 07/21/2023] Open
Abstract
Although the role of the Wnt pathway in colon carcinogenesis has been described previously, it has been recently demonstrated that Wnt signaling originates from highly dynamic nano-assemblies at the plasma membrane. However, little is known regarding the role of oncogenic APC in reshaping Wnt nanodomains. This is noteworthy, because oncogenic APC does not act autonomously and requires activation of Wnt effectors upstream of APC to drive aberrant Wnt signaling. Here, we demonstrate the role of oncogenic APC in increasing plasma membrane free cholesterol and rigidity, thereby modulating Wnt signaling hubs. This results in an overactivation of Wnt signaling in the colon. Finally, using the Drosophila sterol auxotroph model, we demonstrate the unique ability of exogenous free cholesterol to disrupt plasma membrane homeostasis and drive Wnt signaling in a wildtype APC background. Collectively, these findings provide a link between oncogenic APC, loss of plasma membrane homeostasis and CRC development.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Mohamed Mlih
- Department of Cell Biology and Genetics, Texas A&M University, School of Medicine, Bryan, TX, 77807, USA
| | - Venkataramana Thiriveedi
- Department of Medicine, Division of Gastroenterology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Michael L Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Jaileen M Rivera-Rodríguez
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Eunjoo Kim
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, 80045, USA
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Kerstin K Landrock
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Jennifer S Goldsby
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Destiny A Mullens
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Jatin Roper
- Department of Medicine, Division of Gastroenterology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jason Karpac
- Department of Cell Biology and Genetics, Texas A&M University, School of Medicine, Bryan, TX, 77807, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA.
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA.
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA.
- Center for Environmental Health Research, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
22
|
Windfelder AG, Steinbart J, Flögel U, Scherberich J, Kampschulte M, Krombach GA, Vilcinskas A. A quantitative micro-tomographic gut atlas of the lepidopteran model insect Manduca sexta. iScience 2023; 26:106801. [PMID: 37378344 PMCID: PMC10291339 DOI: 10.1016/j.isci.2023.106801] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/26/2023] [Accepted: 04/28/2023] [Indexed: 06/29/2023] Open
Abstract
The tobacco hornworm is used extensively as a model system for ecotoxicology, immunology and gut physiology. Here, we established a micro-computed tomography approach based on the oral application of the clinical contrast agent iodixanol, allowing for a high-resolution quantitative analysis of the Manduca sexta gut. This technique permitted the identification of previously unknown and understudied structures, such as the crop or gastric ceca, and revealed the underlying complexity of the hindgut folding pattern, which is involved in fecal pellet formation. The acquired data enabled the volume rendering of all gut parts, the reliable calculation of their volumes, and the virtual endoscopy of the entire alimentary tract. It can provide information for accurate orientation in histology uses, enable quantitative anatomical phenotyping in three dimensions, and allow the calculation of locally effective midgut concentrations of applied chemicals. This atlas will provide critical insights into the evolution of the alimentary tract in lepidopterans.
Collapse
Affiliation(s)
- Anton G. Windfelder
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- Laboratory of Experimental Radiology, Justus Liebig University Giessen, Giessen, Germany
| | - Jessica Steinbart
- Laboratory of Experimental Radiology, Justus Liebig University Giessen, Giessen, Germany
- Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Jan Scherberich
- Laboratory of Experimental Radiology, Justus Liebig University Giessen, Giessen, Germany
| | - Marian Kampschulte
- Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Germany
| | - Gabriele A. Krombach
- Laboratory of Experimental Radiology, Justus Liebig University Giessen, Giessen, Germany
- Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Germany
| | - Andreas Vilcinskas
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
23
|
Hodge RA, Ghannam M, Edmond E, de la Torre F, D’Alterio C, Kaya NH, Resnik-Docampo M, Reiff T, Jones DL. The septate junction component bark beetle is required for Drosophila intestinal barrier function and homeostasis. iScience 2023; 26:106901. [PMID: 37332603 PMCID: PMC10276166 DOI: 10.1016/j.isci.2023.106901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/19/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Age-related loss of intestinal barrier function has been documented across species, but the causes remain unknown. The intestinal barrier is maintained by tight junctions (TJs) in mammals and septate junctions (SJs) in insects. Specialized TJs/SJs, called tricellular junctions (TCJs), are located at the nexus of three adjacent cells, and we have shown that aging results in changes to TCJs in intestines of adult Drosophila melanogaster. We now demonstrate that localization of the TCJ protein bark beetle (Bark) decreases in aged flies. Depletion of bark from enterocytes in young flies led to hallmarks of intestinal aging and shortened lifespan, whereas depletion of bark in progenitor cells reduced Notch activity, biasing differentiation toward the secretory lineage. Our data implicate Bark in EC maturation and maintenance of intestinal barrier integrity. Understanding the assembly and maintenance of TCJs to ensure barrier integrity may lead to strategies to improve tissue integrity when function is compromised.
Collapse
Affiliation(s)
- Rachel A. Hodge
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mirna Ghannam
- Institute of Genetics, Heinrich-Heine-University, Düsseldorf, Germany
| | - Emma Edmond
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Fernando de la Torre
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cecilia D’Alterio
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nida Hatice Kaya
- Institute of Genetics, Heinrich-Heine-University, Düsseldorf, Germany
| | - Martin Resnik-Docampo
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tobias Reiff
- Institute of Genetics, Heinrich-Heine-University, Düsseldorf, Germany
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, Division of Geriatrics, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Molina-Mateo D, Valderrama BP, Zárate RV, Hidalgo S, Tamayo-Leiva J, Soto A, Guerra S, Arriagada V, Oliva C, Diez B, Campusano JM. Kanamycin treatment in the pre-symptomatic stage of a Drosophila PD model prevents the onset of non-motor alterations. Neuropharmacology 2023; 236:109573. [PMID: 37196855 DOI: 10.1016/j.neuropharm.2023.109573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor alterations, which is preceded by a prodromal stage where non-motor symptoms are observed. Over recent years, it has become evident that this disorder involves other organs that communicate with the brain like the gut. Importantly, the microbial community that lives in the gut plays a key role in this communication, the so-called microbiota-gut-brain axis. Alterations in this axis have been associated to several disorders including PD. Here we proposed that the gut microbiota is different in the presymptomatic stage of a Drosophila model for PD, the Pink1B9 mutant fly, as compared to that observed in control animals. Our results show this is the case: there is basal dysbiosis in mutant animals evidenced by substantial difference in the composition of midgut microbiota in 8-9 days old Pink1B9 mutant flies as compared with control animals. Further, we fed young adult control and mutant flies kanamycin and analyzed motor and non-motor behavioral parameters in these animals. Data show that kanamycin treatment induces the recovery of some of the non-motor parameters altered in the pre-motor stage of the PD fly model, while there is no substantial change in locomotor parameters recorded at this stage. On the other hand, our results show that feeding young animals the antibiotic, results in a long-lasting improvement of locomotion in control flies. Our data support that manipulations of gut microbiota in young animals could have beneficial effects on PD progression and age-dependent motor impairments.
Collapse
Affiliation(s)
- D Molina-Mateo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Centro Interdisciplinario de Neurociencia UC, Pontificia Universidad Católica de Chile, Chile
| | - B P Valderrama
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - R V Zárate
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - S Hidalgo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - J Tamayo-Leiva
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Center for Genome Regulation, Faculty of Science, University of Chile, Santiago, Chile
| | - A Soto
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - S Guerra
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - V Arriagada
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - C Oliva
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - B Diez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Center for Genome Regulation, Faculty of Science, University of Chile, Santiago, Chile; Center for Climate and Resilience Research, University of Chile, Santiago, Chile
| | - J M Campusano
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Centro Interdisciplinario de Neurociencia UC, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
25
|
Salazar AM, Aparicio R, Clark RI, Rera M, Walker DW. Intestinal barrier dysfunction: an evolutionarily conserved hallmark of aging. Dis Model Mech 2023; 16:dmm049969. [PMID: 37144684 PMCID: PMC10184675 DOI: 10.1242/dmm.049969] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
A major challenge in the biology of aging is to understand how specific age-onset pathologies relate to the overall health of the organism. The integrity of the intestinal epithelium is essential for the wellbeing of the organism throughout life. In recent years, intestinal barrier dysfunction has emerged as an evolutionarily conserved feature of aged organisms, as reported in worms, flies, fish, rodents and primates. Moreover, age-onset intestinal barrier dysfunction has been linked to microbial alterations, elevated immune responses, metabolic alterations, systemic health decline and mortality. Here, we provide an overview of these findings. We discuss early work in the Drosophila model that sets the stage for examining the relationship between intestinal barrier integrity and systemic aging, then delve into research in other organisms. An emerging concept, supported by studies in both Drosophila and mice, is that directly targeting intestinal barrier integrity is sufficient to promote longevity. A better understanding of the causes and consequences of age-onset intestinal barrier dysfunction has significant relevance to the development of interventions to promote healthy aging.
Collapse
Affiliation(s)
- Anna M. Salazar
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA 23606, USA
| | - Ricardo Aparicio
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Rebecca I. Clark
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Michael Rera
- Université de Paris, Inserm U1284, Center for Research and Interdisciplinarity, Paris 75004, France
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
Feng P, Wang Y, Zou H, Zhu Q, Ren Y, Shu Q, Su W, Liu W, Hu Y, Li B. The effects of glyphosate exposure on gene transcription and immune function of the silkworm, Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 112:e21990. [PMID: 36537163 DOI: 10.1002/arch.21990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/17/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Glyphosate is a widely used herbicide and crop desiccant. However, whether its extensive use has any effect on the species diversity of nontarget organisms is still unclear. In this study, we used the silkworm, Bombyx mori, as the research subject, and performed RNA sequencing to analyze the transcriptional profile of silkworm midgut after exposure to glyphosate at 2975.20 mg/L (a concentration commonly used at mulberry fields). A total of 125 significantly differentially expressed genes (DEGs) were detected in the midgut of glyphosate-exposed silkworm (q < 0.05), of which 53 were upregulated and 72 were downregulated. Gene ontology enrichment analysis showed that the DEGs were mainly enriched in biological process, cellular component, and molecular function. Kyoto encyclopedia of genes and genomes analysis showed that the differential genes were mainly related to oxidative stress, nutrient metabolism, and immune defense pathways, including oxidative stress-related Cat and Jafrac1, nutrient metabolism-related Fatp and Scpx, and immune-related CYP6AN2, UGT40B4, CTL11, serpin-2, and so forth. Experimental verification showed that glyphosate exposure led to a 4.35-fold increase in the mortality of silkworm after Beauveria bassiana infection, which might be caused by the decreased PO (phenoloxidase) activity and impaired immunity. These results provide evidence for the potential effects of residue glyphosate on the physiological functions of silkworm, and also provide a reference for the biosafety evaluation of glyphosate.
Collapse
Affiliation(s)
- Piao Feng
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yuanfei Wang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Hongbin Zou
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Qingyu Zhu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yuying Ren
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Qilong Shu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Wujie Su
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Wei Liu
- Suzhou Taihu snow silk Co., Ltd, Suzhou, Jiangsu, P.R. China
| | - Yufang Hu
- Suzhou Taihu snow silk Co., Ltd, Suzhou, Jiangsu, P.R. China
| | - Bing Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, P.R. China
- Sericulture Institute of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
27
|
Tang X, Liu N, Qi H, Lin H. Piwi maintains homeostasis in the Drosophila adult intestine. Stem Cell Reports 2023; 18:503-518. [PMID: 36736325 PMCID: PMC9969073 DOI: 10.1016/j.stemcr.2023.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
PIWI genes are well known for their germline but not somatic functions. Here, we report the function of the Drosophila piwi gene in the adult gut, where intestinal stem cells (ISCs) produce enteroendocrine cells and enteroblasts that generate enterocytes. We show that piwi is expressed in ISCs and enteroblasts. Piwi deficiency reduced ISC number, compromised enteroblasts maintenance, and induced apoptosis in enterocytes, but did not affect ISC proliferation and its differentiation to enteroendocrine cells. In addition, deficiency of zygotic but not maternal piwi mildly de-silenced several retrotransposons in the adult gut. Importantly, either piwi mutations or piwi knockdown specifically in ISCs and enteroblasts shortened the Drosophila lifespan, indicating that intestinal piwi contributes to longevity. Finally, our mRNA sequencing data implied that Piwi may achieve its intestinal function by regulating diverse molecular processes involved in metabolism and oxidation-reduction reaction.
Collapse
Affiliation(s)
- Xiongzhuo Tang
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06519, USA.
| | - Na Liu
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Hongying Qi
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Haifan Lin
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
28
|
Kotronarou K, Charalambous A, Evangelou A, Georgiou O, Demetriou A, Apidianakis Y. Dietary Stimuli, Intestinal Bacteria and Peptide Hormones Regulate Female Drosophila Defecation Rate. Metabolites 2023; 13:metabo13020264. [PMID: 36837883 PMCID: PMC9965912 DOI: 10.3390/metabo13020264] [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: 01/20/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Peptide hormones control Drosophila gut motility, but the intestinal stimuli and the gene networks coordinating this trait remain poorly defined. Here, we customized an assay to quantify female Drosophila defecation rate as a proxy of intestinal motility. We found that bacterial infection with the human opportunistic bacterial pathogen Pseudomonas aeruginosa (strain PA14) increases defecation rate in wild-type female flies, and we identified specific bacteria of the fly microbiota able to increase defecation rate. In contrast, dietary stress, imposed by either water-only feeding or high ethanol consumption, decreased defecation rate and the expression of enteroendocrine-produced hormones in the fly midgut, such as Diuretic hormone 31 (Dh31). The decrease in defecation due to dietary stress was proportional to the impact of each stressor on fly survival. Furthermore, we exploited the Drosophila Genetic Reference Panel wild type strain collection and identified strains displaying high and low defecation rates. We calculated the narrow-sense heritability of defecation rate to be 91%, indicating that the genetic variance observed using our assay is mostly additive and polygenic in nature. Accordingly, we performed a genome-wide association (GWA) analysis revealing 17 candidate genes linked to defecation rate. Downregulation of four of them (Pmp70, CG11307, meso18E and mub) in either the midgut enteroendocrine cells or in neurons reduced defecation rate and altered the midgut expression of Dh31, that in turn regulates defecation rate via signaling to the visceral muscle. Hence, microbial and dietary stimuli, and Dh31-controlling genes, regulate defecation rate involving signaling within and among neuronal, enteroendocrine, and visceral muscle cells.
Collapse
|
29
|
Kietz C, Meinander A. Drosophila caspases as guardians of host-microbe interactions. Cell Death Differ 2023; 30:227-236. [PMID: 35810247 PMCID: PMC9950452 DOI: 10.1038/s41418-022-01038-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
An intact cell death machinery is not only crucial for successful embryonic development and tissue homeostasis, but participates also in the defence against pathogens and contributes to a balanced immune response. Centrally involved in the regulation of both cell death and inflammatory immune responses is the evolutionarily conserved family of cysteine proteases named caspases. The Drosophila melanogaster genome encodes for seven caspases, several of which display dual functions, participating in apoptotic signalling and beyond. Among the Drosophila caspases, the caspase-8 homologue Dredd has a well-characterised role in inflammatory signalling activated by bacterial infections, and functions as a driver of NF-κB-mediated immune responses. Regarding the other Drosophila caspases, studies focusing on tissue-specific immune signalling and host-microbe interactions have recently revealed immunoregulatory functions of the initiator caspase Dronc and the effector caspase Drice. The aim of this review is to give an overview of the signalling cascades involved in the Drosophila humoral innate immune response against pathogens and of their caspase-mediated regulation. Furthermore, the apoptotic role of caspases during antibacterial and antiviral immune activation will be discussed.
Collapse
Affiliation(s)
- Christa Kietz
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland.
- InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland.
| |
Collapse
|
30
|
Abstract
The gut epithelia of virtually all animals harbor complex microbial communities that play an important role in maintaining immune and cellular homeostasis. Gut microbiota have evolutionarily adapted to the host gut environment, serving as key regulators of intestinal stem cells to promote a healthy gut barrier and modulate epithelial self-renewal. Disruption of these populations has been associated with inflammatory disorders or cancerous lesions of the intestine. However, the molecular mechanisms controlling gut-microbe interactions are only partially understood due to the high diversity and biologically dynamic nature of these microorganisms. This article reviews the current knowledge on Drosophila gut microbiota and its role in signaling pathways that are crucial for the induction of distinct homeostatic and immune responses. Thanks to the genetic tractability of Drosophila and its cultivable and simple microbiota, this association model offers new efficient tools for investigating the crosstalk between a host and its microbiota while providing a framework for a better understanding of the ecological and evolutionary roles of the microbiome.
Collapse
Affiliation(s)
- Ghada Tafesh-Edwards
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington DC, USA
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington DC, USA
| |
Collapse
|
31
|
Probiotic Bifidobacteria Mitigate the Deleterious Effects of para-Cresol in a Drosophila melanogaster Toxicity Model. mSphere 2022; 7:e0044622. [PMID: 36321825 PMCID: PMC9769938 DOI: 10.1128/msphere.00446-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Renal impairment associated with chronic kidney disease (CKD) causes the buildup of uremic toxins that are deleterious to patient health. Current therapies that manage toxin accumulation in CKD offer an incomplete therapeutic effect against toxins such as para-cresol (p-cresol) and p-cresyl sulfate. Probiotic therapies can exploit the wealth of microbial diversity to reduce toxin accumulation. Using in vitro culture techniques, strains of lactobacilli and bifidobacteria from a 24-strain synbiotic were investigated for their ability to remove p-cresol. Four strains of bifidobacteria internalized p-cresol from the extracellular environment. The oral supplementation of these toxin-clearing probiotics was more protective than control strains in a Drosophila melanogaster toxicity model. Bifidobacterial supplementation was also associated with higher abundance of lactobacilli in the gut microbiota of p-cresol-exposed flies. The present findings suggest that these strains might reduce p-cresol in the gut in addition to increasing the prevalence of other beneficial bacteria, such as lactobacilli, and should be tested clinically to normalize the dysbiotic gut microbiota observed in CKD patients. IMPORTANCE Chronic kidney disease (CKD) affects approximately 10% of the global population and has limited treatment options. The accumulation of gut microbiota-derived uremic toxins, such as para-cresol (p-cresol) and p-cresyl sulfate, is associated with the onset of comorbidities (i.e., atherosclerosis and cognitive disorders) in CKD. Unfortunately, dialysis, the gold standard therapy is unable to remove these toxins from the bloodstream due to their highly protein-bound nature. Some strains of Bifidobacterium have metabolic properties that may be useful in managing uremic toxicity. Using a Drosophila model, the present work highlights why dosing with certain probiotic strains may be clinically useful in CKD management.
Collapse
|
32
|
Chen Z, Wang F, Wen D, Mu R. Exposure to bisphenol A induced oxidative stress, cell death and impaired epithelial homeostasis in the adult Drosophila melanogaster midgut. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114285. [PMID: 36402076 DOI: 10.1016/j.ecoenv.2022.114285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Recently, the chemical compound Bisphenol A (BPA) has been attracting worldwide attention due to its various toxic effects in animals, including reprotoxicity, neurotoxicity, hepatoxicity, and nephrotoxicity. Here, the midgut of adult Drosophila melanogaster (D. melanogaster), an invertebrate model organism, was employed to investigate the gastrointestinal toxicity of BPA in D. melanogaster and explore its underlying mechanisms of action in insects. As a result, exposure of flies to 0.5 mM BPA resulted in a dramatic morphological alteration of D. melanogaster midgut and decrease in survival rates and climbing ability of flies. Further study indicated that BPA induced high levels of oxidative stress in D. melanogaster midgut due to the imbalance between the production of reactive oxygen species and the activities of cellular antioxidant enzymes, including glutathione-S-transferase, catalase and superoxide dismutase. Oxidative stress induced by BPA then caused intestinal epithelial cell death and gut barrier dysfunction and elevated gut permeability, leading to oxidative injury of midgut epithelium. Antioxidant vitamin E alleviated midgut injury induced by BPA. Subsequently, BPA-induced oxidative injury of midgut further stimulated the proliferation of intestinal stem cell (ISC) and ISC-mediated midgut regeneration, but did not alter cell fate determination of ISCs in Drosophila midgut. Meanwhile, activation of Jun N-terminal kinase signal pathway was found to be required for BPA-induced cell death and tissue regeneration in midgut. Collectively, the present study provided additional evidence from an invertebrate model organism that BPA exposure induced gastrointestinal toxicity in D. melanogaster and further extended our understanding of the molecular mechanisms mediating BPA toxicity in insects.
Collapse
Affiliation(s)
- Zhi Chen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| | - Fen Wang
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China
| | - Di Wen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| | - Ren Mu
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| |
Collapse
|
33
|
Xiu M, Wang Y, Yang D, Zhang X, Dai Y, Liu Y, Lin X, Li B, He J. Using Drosophila melanogaster as a suitable platform for drug discovery from natural products in inflammatory bowel disease. Front Pharmacol 2022; 13:1072715. [PMID: 36545307 PMCID: PMC9760693 DOI: 10.3389/fphar.2022.1072715] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/24/2022] [Indexed: 12/07/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and life-treating inflammatory disease that can occur in multiple parts of the human intestine and has become a worldwide problem with a continually increasing incidence. Because of its mild early symptoms, most of them will not attract people's attention and may cause more serious consequences. There is an urgent need for new therapeutics to prevent disease progression. Natural products have a variety of active ingredients, diverse biological activities, and low toxicity or side effects, which are the new options for preventing and treating the intestinal inflammatory diseases. Because of multiple genetic models, less ethical concerns, conserved signaling pathways with mammals, and low maintenance costs, the fruit fly Drosophila melanogaster has become a suitable model for studying mechanism and treatment strategy of IBD. Here, we review the advantages of fly model as screening platform in drug discovery, describe the conserved molecular pathways as therapetic targets for IBD between mammals and flies, dissect the feasibility of Drosophila model in IBD research, and summarize the natural products for IBD treatment using flies. This review comprehensively elaborates that the benefit of flies as a perfact model to evaluate the therapeutic potential of phytochemicals against IBD.
Collapse
Affiliation(s)
- Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China,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 of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Yixuan Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Dan Yang
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xueyan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuting Dai
- College of Public Health, 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 of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Xingyao Lin
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China
| | - Botong Li
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 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, China,Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China,*Correspondence: Jianzheng He,
| |
Collapse
|
34
|
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: 2] [Impact Index Per Article: 1.0] [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
|
35
|
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: 0.5] [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
|
36
|
Rajagopalan K, Christyraj JDS, Chelladurai KS, Gnanaraja JKJS, Christyraj JRSS. Comparative analysis of the survival and regeneration potential of juvenile and matured earthworm, Eudrilus eugeniae, upon in vivo and in vitro maintenance. In Vitro Cell Dev Biol Anim 2022; 58:587-598. [PMID: 35920958 DOI: 10.1007/s11626-022-00706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022]
Abstract
Eudrilus eugeniae is a clitellum-dependent earthworm that requires intact clitellum segments for its survival and regeneration. The present study aims to interconnect the survival and regeneration ability that varies between in vivo and in vitro maintenance upon different sites of amputation. The amputated portion of the worm that possesses intact clitellum (13th-18th segments) survived and had the potential to regenerate, whereas worms with partial or without clitellum segments only survived and were unable to regenerate. Besides segment length and clitellum segments, clitellum factors also determined the survival, blastemal initiation and differentiation potential. The survivability and regeneration potential of worms were augmented upon in vitro maintenance. Notably, the amputated segments (1st-10th segments) and posterior segments of similar length, which usually die within the 4th day in vivo, survived for more than 60 days in vitro but lacked the regeneration ability. On the other hand, the amputated posterior segments (30th to 37th segments) from juvenile worms, maintained in in vitro condition, survived and initiated blastema with multiple buds but lacked the ability to regenerate. Interestingly, the equal half of adult worm blastema that is maintained in in vitro conditions were able to form the blastema-like structure with the help of a unique stick. The anterior blastema failed to retain the regenerative structure but the posterior portion of the amputated blastema, which is also associated with a small portion of the body segment, showed the ability to retain the regenerative structure. Our results conclude that the survivability is enhanced upon in vitro maintenance and this condition favours the adult dedifferentiated blastemal and stem cell-enriched juvenile posterior segments to form a regenerative blastema.
Collapse
Affiliation(s)
- Kamarajan Rajagopalan
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India
| | - Jackson Durairaj Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.
| | - Karthikeyan Subbiahanadar Chelladurai
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.,School of Health Sciences, Purdue University, West Lafayette, USA
| | | | - Johnson Retnaraj Samuel Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to Be University), Chennai, Tamil Nadu, 621306, India.
| |
Collapse
|
37
|
Chongtham A, Yoo JH, Chin TM, Akingbesote ND, Huda A, Marsh JL, Khoshnan A. Gut Bacteria Regulate the Pathogenesis of Huntington's Disease in Drosophila Model. Front Neurosci 2022; 16:902205. [PMID: 35757549 PMCID: PMC9215115 DOI: 10.3389/fnins.2022.902205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Changes in the composition of gut microbiota are implicated in the pathogenesis of several neurodegenerative disorders. Here, we investigated whether gut bacteria affect the progression of Huntington’s disease (HD) in transgenic Drosophila melanogaster (fruit fly) models expressing full-length or N-terminal fragments of human mutant huntingtin (HTT) protein. We find that elimination of commensal gut bacteria by antibiotics reduces the aggregation of amyloidogenic N-terminal fragments of HTT and delays the development of motor defects. Conversely, colonization of HD flies with Escherichia coli (E. coli), a known pathobiont of human gut with links to neurodegeneration and other morbidities, accelerates HTT aggregation, aggravates immobility, and shortens lifespan. Similar to antibiotics, treatment of HD flies with small compounds such as luteolin, a flavone, or crocin a beta-carotenoid, ameliorates disease phenotypes, and promotes survival. Crocin prevents colonization of E. coli in the gut and alters the levels of commensal bacteria, which may be linked to its protective effects. The opposing effects of E. coli and crocin on HTT aggregation, motor defects, and survival in transgenic Drosophila models support the involvement of gut-brain networks in the pathogenesis of HD.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Jung Hyun Yoo
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Theodore M Chin
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Ngozi D Akingbesote
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Ainul Huda
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - J Lawrence Marsh
- Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Ali Khoshnan
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| |
Collapse
|
38
|
Mechanical Forces Govern Interactions of Host Cells with Intracellular Bacterial Pathogens. Microbiol Mol Biol Rev 2022; 86:e0009420. [PMID: 35285720 PMCID: PMC9199418 DOI: 10.1128/mmbr.00094-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To combat infectious diseases, it is important to understand how host cells interact with bacterial pathogens. Signals conveyed from pathogen to host, and vice versa, may be either chemical or mechanical. While the molecular and biochemical basis of host-pathogen interactions has been extensively explored, relatively less is known about mechanical signals and responses in the context of those interactions. Nevertheless, a wide variety of bacterial pathogens appear to have developed mechanisms to alter the cellular biomechanics of their hosts in order to promote their survival and dissemination, and in turn many host responses to infection rely on mechanical alterations in host cells and tissues to limit the spread of infection. In this review, we present recent findings on how mechanical forces generated by host cells can promote or obstruct the dissemination of intracellular bacterial pathogens. In addition, we discuss how in vivo extracellular mechanical signals influence interactions between host cells and intracellular bacterial pathogens. Examples of such signals include shear stresses caused by fluid flow over the surface of cells and variable stiffness of the extracellular matrix on which cells are anchored. We highlight bioengineering-inspired tools and techniques that can be used to measure host cell mechanics during infection. These allow for the interrogation of how mechanical signals can modulate infection alongside biochemical signals. We hope that this review will inspire the microbiology community to embrace those tools in future studies so that host cell biomechanics can be more readily explored in the context of infection studies.
Collapse
|
39
|
Katsaounou K, Nicolaou E, Vogazianos P, Brown C, Stavrou M, Teloni S, Hatzis P, Agapiou A, Fragkou E, Tsiaoussis G, Potamitis G, Zaravinos A, Andreou C, Antoniades A, Shiammas C, Apidianakis Y. Colon Cancer: From Epidemiology to Prevention. Metabolites 2022; 12:metabo12060499. [PMID: 35736432 PMCID: PMC9229931 DOI: 10.3390/metabo12060499] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers affecting humans, with a complex genetic and environmental aetiology. Unlike cancers with known environmental, heritable, or sex-linked causes, sporadic CRC is hard to foresee and has no molecular biomarkers of risk in clinical use. One in twenty CRC cases presents with an established heritable component. The remaining cases are sporadic and associated with partially obscure genetic, epigenetic, regenerative, microbiological, dietary, and lifestyle factors. To tackle this complexity, we should improve the practice of colonoscopy, which is recommended uniformly beyond a certain age, to include an assessment of biomarkers indicative of individual CRC risk. Ideally, such biomarkers will be causal to the disease and potentially modifiable upon dietary or therapeutic interventions. Multi-omics analysis, including transcriptional, epigenetic as well as metagenomic, and metabolomic profiles, are urgently required to provide data for risk analyses. The aim of this article is to provide a perspective on the multifactorial derailment of homeostasis leading to the initiation of CRC, which may be explored via multi-omics and Gut-on-Chip analysis to identify much-needed predictive biomarkers.
Collapse
Affiliation(s)
- Kyriaki Katsaounou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (K.K.); (S.T.)
| | | | - Paris Vogazianos
- Stremble Ventures Ltd., Limassol 4042, Cyprus; (P.V.); (C.B.); (A.A.)
| | - Cameron Brown
- Stremble Ventures Ltd., Limassol 4042, Cyprus; (P.V.); (C.B.); (A.A.)
| | - Marios Stavrou
- Department of Electrical and Computer Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.S.); (C.A.)
| | - Savvas Teloni
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (K.K.); (S.T.)
| | - Pantelis Hatzis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, Vari 16672, Greece;
| | - Agapios Agapiou
- Department of Chemistry, University of Cyprus, Nicosia 2109, Cyprus;
| | | | | | | | - Apostolos Zaravinos
- Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Basic and Translational Cancer Research Center, Nicosia 1516, Cyprus
| | - Chrysafis Andreou
- Department of Electrical and Computer Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.S.); (C.A.)
| | - Athos Antoniades
- Stremble Ventures Ltd., Limassol 4042, Cyprus; (P.V.); (C.B.); (A.A.)
| | | | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus; (K.K.); (S.T.)
- Correspondence:
| |
Collapse
|
40
|
Chandler JA, Innocent LV, Martinez DJ, Huang IL, Yang JL, Eisen MB, Ludington WB. Microbiome-by-ethanol interactions impact Drosophila melanogaster fitness, physiology, and behavior. iScience 2022; 25:104000. [PMID: 35313693 PMCID: PMC8933687 DOI: 10.1016/j.isci.2022.104000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/24/2021] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota can affect how animals respond to ingested toxins, such as ethanol, which is prevalent in the diets of diverse animals and often leads to negative health outcomes in humans. Ethanol is a complex dietary factor because it acts as a toxin, behavioral manipulator, and nutritional source, with both direct effects on the host as well as indirect ones through the microbiome. Here, we developed a model for chronic, non-intoxicating ethanol ingestion in the adult fruit fly, Drosophila melanogaster, and paired this with the tractability of the fly gut microbiota, which can be experimentally removed. We linked numerous physiological, behavioral, and transcriptional variables to fly fitness, including a combination of intestinal barrier integrity, stored triglyceride levels, feeding behavior, and the immunodeficiency pathway. Our results reveal a complex tradeoff between lifespan and fecundity that is microbiome-dependent and modulated by dietary ethanol and feeding behavior.
Collapse
Affiliation(s)
- James Angus Chandler
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Lina Victoria Innocent
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | - Isaac Li Huang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Jane Lani Yang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Michael Bruce Eisen
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - William Basil Ludington
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
41
|
Microbes affect gut epithelial cell composition through immune-dependent regulation of intestinal stem cell differentiation. Cell Rep 2022; 38:110572. [PMID: 35354023 PMCID: PMC9078081 DOI: 10.1016/j.celrep.2022.110572] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/14/2021] [Accepted: 03/03/2022] [Indexed: 12/29/2022] Open
Abstract
Gut microbes play important roles in host physiology; however, the mechanisms underlying their impact remain poorly characterized. Here, we demonstrate that microbes not only influence gut physiology but also alter its epithelial composition. The microbiota and pathogens both influence intestinal stem cell (ISC) differentiation. Intriguingly, while the microbiota promotes ISC differentiation into enterocytes (EC), pathogens stimulate enteroendocrine cell (EE) fate and long-term accumulation of EEs in the midgut epithelium. Importantly, the evolutionarily conserved Drosophila NFKB (Relish) pushes stem cell lineage specification toward ECs by directly regulating differentiation factors. Conversely, the JAK-STAT pathway promotes EE fate in response to infectious damage. We propose a model in which the balance of microbial pattern recognition pathways, such as Imd-Relish, and damage response pathways, such as JAK-STAT, influence ISC differentiation, epithelial composition, and gut physiology.
Collapse
|
42
|
Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 2022; 38:110505. [PMID: 35263602 DOI: 10.1016/j.celrep.2022.110505] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key regulator of metabolism and interacts with the intestinal microbiome. Here, we study the role of the Drosophila intestinal stem cell (ISC)-specific biotin transporter Smvt in midgut homeostasis, infection-induced regeneration, and tumorigenesis. We show that Smvt-transported biotin in ISCs is necessary for ISC mitosis. Smvt deficiency impairs intestinal maintenance, which can be rescued by the human Smvt, encoded by SLC5A6. ISC-specific, Smvt-silenced flies exhibit microbial dysbiosis, whereby the growth of Providencia sneebia, an opportunistic pathogen, is favored. Dysbiosis correlates with increased Nox expression, reactive oxygen species (ROS), and enterocyte apoptosis. Flies acquire biotin from their diet and microbiota. We show that, when dietary biotin is scarce, biotin-producing commensals, e.g., E. coli, can rescue reduced ISC mitosis. Smvt and commensals also control intestinal tumor growth. Our findings suggest that direct modification of the gut microbiome by biotin can serve as an approach for the treatment of dysbiosis-promoted diseases and tumorigenesis control.
Collapse
Affiliation(s)
- Constantina Neophytou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus.
| |
Collapse
|
43
|
Transfer of Human Microbiome to Drosophila Gut Model. Microorganisms 2022; 10:microorganisms10030553. [PMID: 35336128 PMCID: PMC8948740 DOI: 10.3390/microorganisms10030553] [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: 01/10/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 12/10/2022] Open
Abstract
Laboratory animals with human microbiome have increasingly been used to study the role of bacteria and host interaction. Drosophila melanogaster, as a model of microbiota-host interaction with high reproductive efficiency and high availability, has always been lacking studies of interaction with human gut microbiome. In this study, we attempted to use antibiotic therapy and human fecal exposure strategy to transfer the human microbiome to the drosophila. The method includes depleting the original intestinal bacteria using a broad-spectrum antibiotic and then introducing human microorganisms by a diet supplemented with donor’s fecal samples. The sequencing results showed that 80–87.5% of the OTUs (Operational Taxonomic Units) from donor feces were adopted by the recipient drosophila following 30 days of observation. In comparison to females, the male recipient drosophila inherited more microbiota from the donor feces and had significantly increased lifespan as well as improved vertical climbing ability. Furthermore, distinctly differential expression patterns for age and insulin-like signaling-related genes were obtained for the male vs. female recipients. Only the male drosophila offspring acquired the characteristics of the donor fecal microbiota.
Collapse
|
44
|
Wei T, Wu L, Ji X, Gao Y, Xiao G. Ursolic Acid Protects Sodium Dodecyl Sulfate-Induced Drosophila Ulcerative Colitis Model by Inhibiting the JNK Signaling. Antioxidants (Basel) 2022; 11:antiox11020426. [PMID: 35204308 PMCID: PMC8869732 DOI: 10.3390/antiox11020426] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/05/2023] Open
Abstract
Ursolic acid (UA) is a bioactive molecule widely distributed in various fruits and vegetables, which was reported to play a therapeutic role in ulcerative colitis (UC) induced by toxic chemicals. However, the underlying mechanism has not been well clarified in vivo. Here, using a Drosophila UC model induced by sodium dodecyl sulfate (SDS), we investigated the defensive effect of UA on intestinal damage. The results showed that UA could significantly protect Drosophila from the damage caused by SDS exposure. Further, UA alleviated the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA) induced by SDS and upregulated the activities of total superoxide dismutase (T-SOD) and catalase (CAT). Moreover, the proliferation and differentiation of intestine stem cells (ISCs) as well as the excessive activation of the c-Jun N-terminal kinase (JNK)-dependent JAK/STAT signaling pathway induced by SDS were restored by UA. In conclusion, UA prevents intestine injury from toxic compounds by reducing the JNK/JAK/STAT signaling pathway. UA may provide a theoretical basis for functional food or natural medicine development.
Collapse
Affiliation(s)
- Tian Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; (T.W.); (L.W.); (X.J.); (Y.G.)
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Lei Wu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; (T.W.); (L.W.); (X.J.); (Y.G.)
| | - Xiaowen Ji
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; (T.W.); (L.W.); (X.J.); (Y.G.)
| | - Yan Gao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; (T.W.); (L.W.); (X.J.); (Y.G.)
| | - Guiran Xiao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; (T.W.); (L.W.); (X.J.); (Y.G.)
- Correspondence: ; Tel.: +86-177-3022-7689
| |
Collapse
|
45
|
Neophytou C, Pitsouli C. How Gut Microbes Nurture Intestinal Stem Cells: A Drosophila Perspective. Metabolites 2022; 12:169. [PMID: 35208243 PMCID: PMC8878600 DOI: 10.3390/metabo12020169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Host-microbiota interactions are key modulators of host physiology and behavior. Accumulating evidence suggests that the complex interplay between microbiota, diet and the intestine controls host health. Great emphasis has been given on how gut microbes have evolved to harvest energy from the diet to control energy balance, host metabolism and fitness. In addition, many metabolites essential for intestinal homeostasis are mainly derived from gut microbiota and can alleviate nutritional imbalances. However, due to the high complexity of the system, the molecular mechanisms that control host-microbiota mutualism, as well as whether and how microbiota affects host intestinal stem cells (ISCs) remain elusive. Drosophila encompasses a low complexity intestinal microbiome and has recently emerged as a system that might uncover evolutionarily conserved mechanisms of microbiota-derived nutrient ISC regulation. Here, we review recent studies using the Drosophila model that directly link microbiota-derived metabolites and ISC function. This research field provides exciting perspectives for putative future treatments of ISC-related diseases based on monitoring and manipulating intestinal microbiota.
Collapse
Affiliation(s)
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia, Nicosia 2109, Cyprus;
| |
Collapse
|
46
|
Anjum M, Laitila A, Ouwehand AC, Forssten SD. Current Perspectives on Gastrointestinal Models to Assess Probiotic-Pathogen Interactions. Front Microbiol 2022; 13:831455. [PMID: 35173703 PMCID: PMC8841803 DOI: 10.3389/fmicb.2022.831455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
There are different models available that mimic the human intestinal epithelium and are thus available for studying probiotic and pathogen interactions in the gastrointestinal tract. Although, in vivo models make it possible to study the overall effects of a probiotic on a living subject, they cannot always be conducted and there is a general commitment to reduce the use of animal models. Hence, in vitro methods provide a more rapid tool for studying the interaction between probiotics and pathogens; as well as being ethically superior, faster, and less expensive. The in vitro models are represented by less complex traditional models, standard 2D models compromised of culture plates as well as Transwell inserts, and newer 3D models like organoids, enteroids, as well as organ-on-a-chip. The optimal model selected depends on the research question. Properly designed in vitro and/or in vivo studies are needed to examine the mechanism(s) of action of probiotics on pathogens to obtain physiologically relevant results.
Collapse
Affiliation(s)
| | | | | | - Sofia D. Forssten
- International Flavors and Fragrances, Health and Biosciences, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
47
|
Eickelberg V, Lüersen K, Staats S, Rimbach G. Phenotyping of Drosophila Melanogaster-A Nutritional Perspective. Biomolecules 2022; 12:221. [PMID: 35204721 PMCID: PMC8961528 DOI: 10.3390/biom12020221] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
The model organism Drosophila melanogaster was increasingly applied in nutrition research in recent years. A range of methods are available for the phenotyping of D. melanogaster, which are outlined in the first part of this review. The methods include determinations of body weight, body composition, food intake, lifespan, locomotor activity, reproductive capacity and stress tolerance. In the second part, the practical application of the phenotyping of flies is demonstrated via a discussion of obese phenotypes in response to high-sugar diet (HSD) and high-fat diet (HFD) feeding. HSD feeding and HFD feeding are dietary interventions that lead to an increase in fat storage and affect carbohydrate-insulin homeostasis, lifespan, locomotor activity, reproductive capacity and stress tolerance. Furthermore, studies regarding the impacts of HSD and HFD on the transcriptome and metabolome of D. melanogaster are important for relating phenotypic changes to underlying molecular mechanisms. Overall, D. melanogaster was demonstrated to be a valuable model organism with which to examine the pathogeneses and underlying molecular mechanisms of common chronic metabolic diseases in a nutritional context.
Collapse
Affiliation(s)
- Virginia Eickelberg
- Department of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany; (K.L.); (S.S.); (G.R.)
| | | | | | | |
Collapse
|
48
|
Thangadurai S, Bajgiran M, Manickam S, Mohana-Kumaran N, Azzam G. CTP synthase: the hissing of the cellular serpent. Histochem Cell Biol 2022; 158:517-534. [PMID: 35881195 PMCID: PMC9314535 DOI: 10.1007/s00418-022-02133-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 12/24/2022]
Abstract
CTP biosynthesis is carried out by two pathways: salvage and de novo. CTPsyn catalyzes the latter. The study of CTPsyn activity in mammalian cells began in the 1970s, and various fascinating discoveries were made regarding the role of CTPsyn in cancer and development. However, its ability to fit into a cellular serpent-like structure, termed 'cytoophidia,' was only discovered a decade ago by three independent groups of scientists. Although the self-assembly of CTPsyn into a filamentous structure is evolutionarily conserved, the enzyme activity upon this self-assembly varies in different species. CTPsyn is required for cellular development and homeostasis. Changes in the expression of CTPsyn cause developmental changes in Drosophila melanogaster. A high level of CTPsyn activity and formation of cytoophidia are often observed in rapidly proliferating cells such as in stem and cancer cells. Meanwhile, the deficiency of CTPsyn causes severe immunodeficiency leading to immunocompromised diseases caused by bacteria, viruses, and parasites, making CTPsyn an attractive therapeutic target. Here, we provide an overview of the role of CTPsyn in cellular and disease perspectives along with its potential as a drug target.
Collapse
Affiliation(s)
- Shallinie Thangadurai
- grid.11875.3a0000 0001 2294 3534School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Morteza Bajgiran
- grid.11875.3a0000 0001 2294 3534School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Sharvin Manickam
- grid.11875.3a0000 0001 2294 3534School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Nethia Mohana-Kumaran
- grid.11875.3a0000 0001 2294 3534School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Ghows Azzam
- grid.11875.3a0000 0001 2294 3534School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia ,grid.454125.3Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000 Kajang, Selangor Malaysia
| |
Collapse
|
49
|
Kietz C, Mohan AK, Pollari V, Tuominen IE, Ribeiro PS, Meier P, Meinander A. Drice restrains Diap2-mediated inflammatory signalling and intestinal inflammation. Cell Death Differ 2022; 29:28-39. [PMID: 34262145 PMCID: PMC8738736 DOI: 10.1038/s41418-021-00832-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The Drosophila IAP protein, Diap2, is a key mediator of NF-κB signalling and innate immune responses. Diap2 is required for both local immune activation, taking place in the epithelial cells of the gut and trachea, and for mounting systemic immune responses in the cells of the fat body. We have found that transgenic expression of Diap2 leads to a spontaneous induction of NF-κB target genes, inducing chronic inflammation in the Drosophila midgut, but not in the fat body. Drice is a Drosophila effector caspase known to interact and form a stable complex with Diap2. We have found that this complex formation induces its subsequent degradation, thereby regulating the amount of Diap2 driving NF-κB signalling in the intestine. Concordantly, loss of Drice activity leads to accumulation of Diap2 and to chronic intestinal inflammation. Interestingly, Drice does not interfere with pathogen-induced signalling, suggesting that it protects from immune responses induced by resident microbes. Accordingly, no inflammation was detected in transgenic Diap2 flies and Drice-mutant flies reared in axenic conditions. Hence, we show that Drice, by restraining Diap2, halts unwanted inflammatory signalling in the intestine.
Collapse
Affiliation(s)
- Christa Kietz
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Aravind K Mohan
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Vilma Pollari
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Ida-Emma Tuominen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland.
| |
Collapse
|
50
|
Snow JW. Nosema apis and N. ceranae Infection in Honey bees: A Model for Host-Pathogen Interactions in Insects. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 114:153-177. [PMID: 35544003 DOI: 10.1007/978-3-030-93306-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
There has been increased focus on the role of microbial attack as a potential cause of recent declines in the health of the western honey bee, Apis mellifera. The Nosema species, N. apis and N. ceranae, are microsporidian parasites that are pathogenic to honey bees, and infection by these species has been implicated as a key factor in honey bee losses. Honey bees infected with both Nosema spp. display significant changes in their biology at the cellular, tissue, and organismal levels impacting host metabolism, immune function, physiology, and behavior. Infected individuals lead to colony dysfunction and can contribute to colony disease in some circumstances. The means through which parasite growth and tissue pathology in the midgut lead to the dramatic physiological and behavioral changes at the organismal level are only partially understood. In addition, we possess only a limited appreciation of the elements of the host environment that impact pathogen growth and development. Critical for answering these questions is a mechanistic understanding of the host and pathogen machinery responsible for host-pathogen interactions. A number of approaches are already being used to elucidate these mechanisms, and promising new tools may allow for gain- and loss-of-function experiments to accelerate future progress.
Collapse
|