1
|
Moreira DR, de Souza THS, Galhardo D, Figueira CL, Baulli SC, da Silva BG, das Chagas F, Oliveira JWS, Rocha JS, de Souza Khatlab A, Gasparino E, de Alencar Arnaut de Toledo V, Gigliolli AAS, Ruvolo-Takasusuki MCC. Exposure of Apis mellifera (Hymenoptera: Apidae) colonies to imidacloprid impairs larval development, promotes oxidative stress in pupae, and induces changes in the midgut of adult bees. Biol Res 2025; 58:5. [PMID: 39833873 PMCID: PMC11748266 DOI: 10.1186/s40659-024-00571-5] [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: 12/08/2023] [Accepted: 11/07/2024] [Indexed: 01/22/2025] Open
Abstract
Bees are essential pollinators that contribute to maintaining biodiversity and increasing agricultural production. However, by foraging on agricultural crops, bees may become contaminated with compounds used for pest control. In this study, we exposed bee (Apis mellifera L.) colonies to the insecticide imidacloprid (IMD) under field conditions to assess the occurrence of oxidative stress in larvae and pupae and investigate morphological changes in the fat body and midgut of larvae and midgut of adult bees. The apiary area was divided into three groups: control, commercial formulation containing IMD (Evidence® 700WG) (IMDCF), and IMD active ingredient (Sigma-Aldrich) (IMDAI). Treatment groups were fed syrup containing 1 µg L-1 IMD, whereas the control group was fed syrup only. Compared with the control, larvae exposed to IMDCF or IMDAI for 42 days exhibited morphological changes in the external body, midgut, and fat body. The midgut of adult bees contaminated with IMDCF showed only structural remnants of the peritrophic membrane and absence of regenerative cell nests. Oxidative stress analyses revealed that IMDCF-exposed larvae had higher nitrite and carbonylated protein contents and lower catalase and superoxide dismutase activity than control individuals. In pupae, IMDAI decreased catalase activity while increasing superoxide dismutase activity. These findings indicate that IMD has the potential to significantly impact the development of bees and their colonies by disrupting vital organs responsible for normal physiological functioning and overall activities of individuals. Oxidative stress, which was detected at different stages of bee development, may induce lipid, protein, and DNA oxidation, leading to cell death.
Collapse
Affiliation(s)
- Daiani Rodrigues Moreira
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | | | - Douglas Galhardo
- Department of Animal Science, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Cinthia Leão Figueira
- Department of Animal Science, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Samara Calvi Baulli
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Breno Gabriel da Silva
- Department of Exact Sciences, Escola Superior de Agricultura "Luiz de Queiroz" - University of São Paulo, Piracicaba, São Paulo, 13418-900, Brazil
| | - Francieli das Chagas
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
- Centre of Molecular Biodiversity Research, Leibniz Institute for the Analysis of Biodiversity Change, Zoological Research Museum Alexander Koenig, Adenauerallee 160, 53113, Bonn, Germany
| | | | - Jean Samel Rocha
- Department of Animal Science, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | | | - Eliane Gasparino
- Department of Animal Science, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | | | | | | |
Collapse
|
2
|
Monastirioti M, Koltsaki I, Pitsidianaki I, Skafida E, Batsiotos N, Delidakis C. Notch-Dependent Expression of the Drosophila Hey Gene Is Supported by a Pair of Enhancers with Overlapping Activities. Genes (Basel) 2024; 15:1071. [PMID: 39202431 PMCID: PMC11353301 DOI: 10.3390/genes15081071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/09/2024] [Accepted: 08/10/2024] [Indexed: 09/03/2024] Open
Abstract
Drosophila Hey is a basic helix-loop-helix-orange (bHLH-O) protein with an important role in the establishment of distinct identities of postmitotic cells. We have previously identified Hey as a transcriptional target and effector of Notch signalling during the asymmetric division of neuronal progenitors, generating neurons of two types, and we have shown that Notch-dependent expression of Hey also marks a subpopulation of the newborn enteroendocrine (EE) cells in the midgut primordium of the embryo. Here, we investigate the transcriptional regulation of Hey in neuronal and intestinal tissues. We isolated two genomic regions upstream of the promoter (HeyUP) and in the second intron (HeyIN2) of the Hey gene, based on the presence of binding motifs for Su(H), the transcription factor that mediates Notch activity. We found that both regions can direct the overlapping expression patterns of reporter transgenes recapitulating endogenous Hey expression. Moreover, we showed that while HeyIN2 represents a Notch-dependent enhancer, HeyUP confers both Notch-dependent and independent transcriptional regulation. We induced mutations that removed the Su(H) binding motifs in either region and then studied the enhancer functionality in the respective Hey mutant lines. Our results provide direct evidence that although both enhancers support Notch-dependent regulation of the Hey gene, their role is redundant, as a Hey loss-of-function lethal phenotype is observed only after deletion of all their Su(H) binding motifs by CRISPR/Cas9.
Collapse
Affiliation(s)
- Maria Monastirioti
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
| | - Ioanna Koltsaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ioanna Pitsidianaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece
- Department of Cell and Developmental Biology, University College London (UCL), London WC1E 6BT, UK
| | - Emilia Skafida
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Foundation Saint Lucia, Rome and School of Medicine and Surgery, University of Milano-Bicocca (UniMiB), 20900 Monza, Italy
| | - Nikolaos Batsiotos
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece
- Evotec SE, 22419 Hamburg, Germany
| | - Christos Delidakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), 70013 Heraklion, Greece; (I.K.); (I.P.); (E.S.); (N.B.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
3
|
Nutrient Sensing via Gut in Drosophila melanogaster. Int J Mol Sci 2022; 23:ijms23052694. [PMID: 35269834 PMCID: PMC8910450 DOI: 10.3390/ijms23052694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Nutrient-sensing mechanisms in animals' sense available nutrients to generate a physiological regulatory response involving absorption, digestion, and regulation of food intake and to maintain glucose and energy homeostasis. During nutrient sensing via the gastrointestinal tract, nutrients interact with receptors on the enteroendocrine cells in the gut, which in return respond by secreting various hormones. Sensing of nutrients by the gut plays a critical role in transmitting food-related signals to the brain and other tissues informing the composition of ingested food to digestive processes. These signals modulate feeding behaviors, food intake, metabolism, insulin secretion, and energy balance. The increasing significance of fly genetics with the availability of a vast toolbox for studying physiological function, expression of chemosensory receptors, and monitoring the gene expression in specific cells of the intestine makes the fly gut the most useful tissue for studying the nutrient-sensing mechanisms. In this review, we emphasize on the role of Drosophila gut in nutrient-sensing to maintain metabolic homeostasis and gut-brain cross talk using endocrine and neuronal signaling pathways stimulated by internal state or the consumption of various dietary nutrients. Overall, this review will be useful in understanding the post-ingestive nutrient-sensing mechanisms having a physiological and pathological impact on health and diseases.
Collapse
|
4
|
Jahnes BC, Poudel K, Staats AM, Sabree ZL. Microbial colonization promotes model cockroach gut tissue growth and development. JOURNAL OF INSECT PHYSIOLOGY 2021; 133:104274. [PMID: 34216600 DOI: 10.1016/j.jinsphys.2021.104274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Digestive tissues are essential for diet processing and nutrient accessibility, especially in omnivores, and these functions occur despite and in collaboration with dynamic microbial communities that reside within and upon these tissues. Prolonged host development and reduced digestive tissue sizes have been observed in germ-free animals, and normal host phenotypes were recovered following the re-introduction of typical gut microbiomes via coprophagy. RESULTS High-resolution histological analyses of Periplaneta americana cockroach digestive tissues revealed that total prevention of microbial colonization of the gut had severe impacts on the growth and development of gut tissues, especially the posterior midgut and anterior hindgut subcompartments that are expected to be colonized and inhabited by the greatest number of bacteria. Juveniles that were briefly exposed to normal gut microbiota exhibited a partial gut morphological recovery, suggesting that a single inoculation was insufficient. These data highlight gut microbiota as integral to normal growth and development of tissues they are in direct contact with and, more broadly, the organism in which they reside. CONCLUSIONS We draw on these data, host life history traits (i.e. multigenerational cohousing, molting, and filial coprophagy and exuvia feeding), and previous studies to suggest a host developmental model in which gut tissues reflect a conflict-collaboration dynamic where 1) nutrient-absorptive anterior midgut tissues are in competition with transient and resident bacteria for easily assimilable dietary nutrients and whose growth is least-affected by the presence of gut bacteria and 2) posterior midgut, anterior hindgut, and to a lesser degree, posterior hindgut tissues are significantly impacted by gut bacterial presence because they are occupied by the greatest number of bacteria and the host is relying upon, and thus collaborating with, them to assist with complex polysaccharide catabolism processing and nutrient provisioning (i.e. short-chain fatty acids).
Collapse
Affiliation(s)
- Benjamin C Jahnes
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | - Keyshap Poudel
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH, USA
| | - Amelia M Staats
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | - Zakee L Sabree
- Department of Microbiology, Ohio State University, Columbus, OH, USA; Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Zhao H, Shi L, Kong R, Li Z, Liu F, Zhao H, Li Z. Autophagy induction in tumor surrounding cells promotes tumor growth in adult Drosophila intestines. Dev Biol 2021; 476:294-307. [PMID: 33940033 DOI: 10.1016/j.ydbio.2021.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
During tumorigenesis, tumor cells interact intimately with their surrounding cells (microenvironment) for their growth and progression. However, the roles of tumor microenvironment in tumor development and progression are not fully understood. Here, using an established benign tumor model in adult Drosophila intestines, we find that non-cell autonomous autophagy (NAA) is induced in tumor surrounding neighbor cells. Tumor growth can be significantly suppressed by genetic ablation of autophagy induction in tumor neighboring cells, indicating that tumor neighboring cells act as tumor microenvironment to promote tumor growth. Autophagy in tumor neighboring cells is induced downstream of elevated ROS and activated JNK signaling in tumor cells. Interestingly, we find that active transport of nutrients, such as amino acids, from tumor neighboring cells sustains tumor growth, and increasing nutrient availability could significantly restore tumor growth. Together, these data demonstrate that tumor cells take advantage of their surrounding normal neighbor cells as nutrient sources through NAA to meet their high metabolic demand for growth and progression. Thus we provide insights into our understanding of the mechanisms underlying the interaction between tumor cells and their microenvironment in tumor development.
Collapse
Affiliation(s)
- Hang Zhao
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Lin Shi
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Ruiyan Kong
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Zhengran Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Fuli Liu
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Huiqing Zhao
- College of Life Sciences, Capital Normal University, Beijing, 100048, China
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China.
| |
Collapse
|
6
|
Ferguson M, Petkau K, Shin M, Galenza A, Fast D, Foley E. Differential effects of commensal bacteria on progenitor cell adhesion, division symmetry and tumorigenesis in the Drosophila intestine. Development 2021; 148:dev.186106. [DOI: 10.1242/dev.186106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Microbial factors influence homeostatic and oncogenic growth in the intestinal epithelium. However, we know little about immediate effects of commensal bacteria on stem cell division programs. In this study, we examined the effects of commensal Lactobacillus species on homeostatic and tumorigenic stem cell proliferation in the female Drosophila intestine. We identified Lactobacillus brevis as a potent stimulator of stem cell divisions. In a wild-type midgut, L.brevis activates growth regulatory pathways that drive stem cell divisions. In a Notch-deficient background, L.brevis-mediated proliferation causes rapid expansion of mutant progenitors, leading to accumulation of large, multi-layered tumors throughout the midgut. Mechanistically, we showed that L.brevis disrupts expression and subcellular distribution of progenitor cell integrins, supporting symmetric divisions that expand intestinal stem cell populations. Collectively, our data emphasize the impact of commensal microbes on division and maintenance of the intestinal progenitor compartment.
Collapse
Affiliation(s)
- Meghan Ferguson
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Kristina Petkau
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Anthony Galenza
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - David Fast
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
7
|
Swevers L, Denecke S, Vogelsang K, Geibel S, Vontas J. Can the mammalian organoid technology be applied to the insect gut? PEST MANAGEMENT SCIENCE 2021; 77:55-63. [PMID: 32865304 DOI: 10.1002/ps.6067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
Mammalian intestinal organoids are multicellular structures that closely resemble the structure of the intestinal epithelium and can be generated in vitro from intestinal stem cells under appropriate culture conditions. This technology has transformed pharmaceutical research and drug development in human medicine. For the insect gut, no biotechnological platform equivalent to organoid cultures has been described yet. Comparison of the regulation of intestinal homeostasis and growth between insects and mammals has revealed significant similarities but also important differences. In contrast to mammals, the differentiation potential of available insect cell lines is limited and can not be exploited for in vitro permeability assays to measure the uptake of insecticides. The successful development of in vitro models could be a result of the emergence of molecular mechanisms of self-organization and signaling in the intestine that are unique to mammals. It is nevertheless considered that the technology gap is a consequence of vast differences in knowledge, particularly with respect to culture conditions that maintain the differentation potential of insect midgut cells. From the viewpoint of pest control, advanced in vitro models of the insect midgut would be very desirable because of its key barrier function for orally ingested insecticides with hemolymphatic target and its role in insecticide resistance. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences & Applications, National Centre for Scientific Research "Demokritos", Agia Paraskevi, 15341, Greece
| | - Shane Denecke
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | | | - Sven Geibel
- Bayer AG, Crop Science Devision, R&D Pest Control, Monheim, Germany
| | - John Vontas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Pesticide Science Lab, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
8
|
Zhang Q, Hao LC, Hong Y. Exposure evaluation of diisononyl phthalate in the adults of Drosophila melanogaster: Potential risks in fertility, lifespan, behavior, and modes of action. Comp Biochem Physiol C Toxicol Pharmacol 2020; 238:108847. [PMID: 32781294 DOI: 10.1016/j.cbpc.2020.108847] [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: 05/05/2020] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 11/19/2022]
Abstract
Diisononyl phthalate (DINP) as a phthalate plasticizer is widely used in daily life and production, which shows endocrine disruption effects and has several adverse effects on the normal physiological function. Here, the effects of DINP (0.1%, 0.2%, 0.5%, and 1.0%) (v/v) on the fertility, lifespan, climbing behavior, anti-starvation ability of Drosophila melanogaster and the potential modes of action were investigated. The results showed that DINP impaired fertility in a dose-dependent manner and smaller ovarian volume, lower hatching rate, and fewer offspring was observed at higher concentrations. The effect of DINP on the lifespan showed gender-specific, and mortality was increased after exposure above 0.2% DINP. The climbing ability increased at 0.1% DINP compared with the vehicle group, while it manifested a dose-dependent decrease at higher concentrations. The anti-starvation ability exhibited hormesis after short-term culture and reduced as culture time extending. By measuring the redox status (catalase (CAT) and reactive oxygen species (ROS)) of adult flies after two exposure methods, it was found that DINP induced redox instability, which may explain the above effects at the molecular level. This study provides data to support a comprehensive analysis of DINP potential toxicity and to guide its rational use and management better.
Collapse
Affiliation(s)
- Qing Zhang
- Beijing Key Lab for Source Control Technology of Water Pollution, College of Environmental Science and Engineering, Beijing Forestry University, Beijing 100083, China
| | - Li-Chong Hao
- Beijing Key Lab for Source Control Technology of Water Pollution, College of Environmental Science and Engineering, Beijing Forestry University, Beijing 100083, China
| | - Yu Hong
- Beijing Key Lab for Source Control Technology of Water Pollution, College of Environmental Science and Engineering, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
9
|
Hall MJR, Martín-Vega D. Visualization of insect metamorphosis. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190071. [PMID: 31438819 DOI: 10.1098/rstb.2019.0071] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Metamorphosis and, in particular, holometaboly, the development of organisms through a series of discrete stages (egg, larva, pupa, adult) that hardly resemble one another but are finely adapted to specific roles in the life cycle of the organism, has fascinated and mystified humans throughout history. However, it can be difficult to visualize the dramatic changes that occur during holometaboly without destructive sampling, traditionally through histology. However, advances in imaging technologies developed mainly for medical sciences have been applied to studies of insect metamorphosis over the past couple of decades. These include micro-computed tomography, magnetic resonance imaging and optical coherence tomography. A major advantage of these techniques is that they are rapid and non-destructive, enabling virtual dissection of an organism in any plane by anyone who has access to the image files and the necessary software. They can also be applied in some cases to visualize metamorphosis in vivo, including the periods of most rapid and dramatic morphological change. This review focusses on visualizing the intra-puparial holometabolous metamorphosis of cyclorraphous flies (Diptera), including the primary model organism for all genetic investigations, Drosophila melanogaster, and the blow flies of medical, veterinary and forensic importance, but also discusses similar studies on other insect orders. This article is part of the theme issue 'The evolution of complete metamorphosis'.
Collapse
Affiliation(s)
- Martin J R Hall
- Department of Life Sciences, Natural History Museum, Cromwell Road, London SW7 5BD, UK
| | - Daniel Martín-Vega
- Department of Life Sciences, Natural History Museum, Cromwell Road, London SW7 5BD, UK.,Department of Life Sciences, University of Alcalá, Alcalá de Henares, Spain
| |
Collapse
|
10
|
Galenza A, Foley E. Immunometabolism: Insights from the Drosophila model. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 94:22-34. [PMID: 30684503 DOI: 10.1016/j.dci.2019.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
Multicellular organisms inhabit an environment that includes a mix of essential nutrients and large numbers of potentially harmful microbes. Germline-encoded receptors scan the environment for microbe associated molecular patterns, and, upon engagement, activate powerful defenses to protect the host from infection. At the same time, digestive enzymes and transporter molecules sieve through ingested material for building blocks and energy sources necessary for survival, growth, and reproduction. We tend to view immune responses as a potent array of destructive forces that overwhelm potentially harmful agents. In contrast, we view metabolic processes as essential, constructive elements in the maintenance and propagation of life. However, there is considerable evidence of functional overlap between the two processes, and disruptions to one frequently modify outputs of the other. Studies of immunometabolism, or interactions between immunity and metabolism, have increased in prominence with the discovery of inflammatory components to metabolic diseases such as type two diabetes. In this review, we will focus on contributions of studies with the fruit fly, Drosophila melanogaster, to our understanding of immunometabolism. Drosophila is widely used to study immune signaling, and to understand the regulation of metabolism in vivo, and this insect has considerable potential as a tool to build our understanding of the molecular and cellular bridges that connect immune and metabolic pathways.
Collapse
Affiliation(s)
- Anthony Galenza
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada.
| |
Collapse
|
11
|
Racing to Stay Put: How Resident Microbiota Stimulate Intestinal Epithelial Cell Proliferation. CURRENT PATHOBIOLOGY REPORTS 2018. [DOI: 10.1007/s40139-018-0163-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Zhang L, Turner B, Ribbeck K, Ten Hagen KG. Loss of the mucosal barrier alters the progenitor cell niche via Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. J Biol Chem 2017; 292:21231-21242. [PMID: 29127201 PMCID: PMC5766965 DOI: 10.1074/jbc.m117.809848] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/23/2017] [Indexed: 01/09/2023] Open
Abstract
The mucous barrier of our digestive tract is the first line of defense against pathogens and damage. Disruptions in this barrier are associated with diseases such as Crohn's disease, colitis, and colon cancer, but mechanistic insights into these processes and diseases are limited. We have previously shown that loss of a conserved O-glycosyltransferase (PGANT4) in Drosophila results in aberrant secretion of components of the peritrophic/mucous membrane in the larval digestive tract. Here, we show that loss of PGANT4 disrupts the mucosal barrier, resulting in epithelial expression of the IL-6-like cytokine Upd3, leading to activation of JAK/STAT signaling, differentiation of cells that form the progenitor cell niche, and abnormal proliferation of progenitor cells. This niche disruption could be recapitulated by overexpressing upd3 and rescued by deleting upd3, highlighting a crucial role for this cytokine. Moreover, niche integrity and cell proliferation in pgant4-deficient animals could be rescued by overexpression of the conserved cargo receptor Tango1 and partially rescued by supplementation with exogenous mucins or treatment with antibiotics. Our findings help elucidate the paracrine signaling events activated by a compromised mucosal barrier and provide a novel in vivo screening platform for mucin mimetics and other strategies to treat diseases of the oral mucosa and digestive tract.
Collapse
Affiliation(s)
- Liping Zhang
- From the Developmental Glycobiology Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892-4370 and
| | - Bradley Turner
- the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Katharina Ribbeck
- the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Kelly G Ten Hagen
- From the Developmental Glycobiology Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892-4370 and
| |
Collapse
|
13
|
Jones TA, Hernandez DZ, Wong ZC, Wandler AM, Guillemin K. The bacterial virulence factor CagA induces microbial dysbiosis that contributes to excessive epithelial cell proliferation in the Drosophila gut. PLoS Pathog 2017; 13:e1006631. [PMID: 29049360 PMCID: PMC5648253 DOI: 10.1371/journal.ppat.1006631] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota facilitate many aspects of human health and development, but dysbiotic microbiota can promote hyperplasia and inflammation and contribute to human diseases such as cancer. Human patients infected with the gastric cancer-causing bacterium Helicobacter pylori have altered microbiota; however, whether dysbiosis contributes to disease in this case is unknown. Many H. pylori human disease phenotypes are associated with a potent virulence protein, CagA, which is translocated into host epithelial cells where it alters cell polarity and manipulates host-signaling pathways to promote disease. We hypothesized that CagA alone could contribute to H. pylori pathogenesis by inducing microbial dysbiosis that promotes disease. Here we use a transgenic Drosophila model of CagA expression to genetically disentangle the effects of the virulence protein CagA from that of H. pylori infection. We found that expression of CagA within Drosophila intestinal stem cells promotes excess cell proliferation and is sufficient to alter host microbiota. Rearing CagA transgenic flies germ-free revealed that the dysbiotic microbiota contributes to cell proliferation phenotypes and also elicits expression of innate immune components, Diptericin and Duox. Further investigations revealed interspecies interactions are required for this dysbiotic CagA-dependent microbiota to promote proliferation in CagA transgenic and healthy control Drosophila. Our model establishes that CagA can alter gut microbiota and exacerbate cell proliferation and immune phenotypes previously attributed to H. pylori infection. This work provides valuable new insights into the mechanisms by which interactions between a specific virulence factor and the resident microbiota can contribute to the development and progression of disease. Microbial communities in the gut, termed microbiota are important for human health, and when altered can sometimes promote disease. Infections, such as with the cancer-causing bacterium Helicobacter pylori, can cause altered gut microbiota, but why these alterations occur and whether the altered communities contribute to disease remain unknown. Here, we use Drosophila expressing the H. pylori disease-causing protein CagA, to model this virulence factor’s effect on host pathology and microbiota. We found that expression of CagA in the Drosophila gut causes excessive cell proliferation and immune activation, hallmarks of H. pylori infection. Notably, these traits did not occur when flies were reared in the absence of microbes. Further examination reveals that CagA-expressing flies have an altered gut microbial community that is sufficient to promote cell proliferation even in normal flies. This proliferative activity required the presence of two interacting bacteria, illustrating a new model for disease-promoting microbiota. This work demonstrates how a bacterial protein can cause disease indirectly through altering the microbial ecology of the host, and it suggests future treatments for infections that rely on manipulating the microbiota to mitigate disease pathology.
Collapse
Affiliation(s)
- Tiffani Alvey Jones
- Institute of Molecular Biology, University of Oregon, Eugene, OR, United States of America
| | - Diane Z. Hernandez
- Institute of Molecular Biology, University of Oregon, Eugene, OR, United States of America
| | - Zoë C. Wong
- Institute of Molecular Biology, University of Oregon, Eugene, OR, United States of America
| | - Anica M. Wandler
- Institute of Molecular Biology, University of Oregon, Eugene, OR, United States of America
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR, United States of America
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
14
|
Abstract
Spinal cord injury (SCI) represents one of the most complicated and heterogeneous pathological processes of central nervous system (CNS) impairments, which is still beyond functional regeneration. Transplantation of mesenchymal stem cells (MSCs) has been shown to promote the repair of the injured spinal cord tissues in animal models, and therefore, there is much interest in the clinical use of these cells. However, many questions which are essential to improve the therapy effects remain unanswered. For instance, the functional roles and related molecular regulatory mechanisms of MSCs in vivo are not yet completely determined. It is important for transplanted cells to migrate into the injured tissue, to survive and undergo neural differentiation, or to play neural protection roles by various mechanisms after SCI. In this review, we will focus on some of the recent knowledge about the biological behavior and function of MSCs in SCI. Meanwhile, we highlight the function of biomaterials to direct the behavior of MSCs based on our series of work on silk fibroin biomaterials and attempt to emphasize combinational strategies such as tissue engineering for functional improvement of SCI.
Collapse
|
15
|
Petkau K, Fast D, Duggal A, Foley E. Comparative evaluation of the genomes of three common Drosophila-associated bacteria. Biol Open 2016; 5:1305-16. [PMID: 27493201 PMCID: PMC5051641 DOI: 10.1242/bio.017673] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Drosophila melanogaster is an excellent model to explore the molecular exchanges that occur between an animal intestine and associated microbes. Previous studies in Drosophila uncovered a sophisticated web of host responses to intestinal bacteria. The outcomes of these responses define critical events in the host, such as the establishment of immune responses, access to nutrients, and the rate of larval development. Despite our steady march towards illuminating the host machinery that responds to bacterial presence in the gut, there are significant gaps in our understanding of the microbial products that influence bacterial association with a fly host. We sequenced and characterized the genomes of three common Drosophila-associated microbes: Lactobacillus plantarum, Lactobacillus brevis and Acetobacter pasteurianus. For each species, we compared the genomes of Drosophila-associated strains to the genomes of strains isolated from alternative sources. We found that environmental Lactobacillus strains readily associated with adult Drosophila and were similar to fly isolates in terms of genome organization. In contrast, we identified a strain of A. pasteurianus that apparently fails to associate with adult Drosophila due to an inability to grow on fly nutrient food. Comparisons between association competent and incompetent A. pasteurianus strains identified a short list of candidate genes that may contribute to survival on fly medium. Many of the gene products unique to fly-associated strains have established roles in the stabilization of host-microbe interactions. These data add to a growing body of literature that examines the microbial perspective of host-microbe relationships. Summary: We examined the genomes of Drosophila-associated bacteria to identify factors that allow survival within the host. These preliminary studies may point at bacterial products that influence host health.
Collapse
Affiliation(s)
- Kristina Petkau
- Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton AB, T6G 2E1 Canada
| | - David Fast
- Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton AB, T6G 2E1 Canada
| | - Aashna Duggal
- Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton AB, T6G 2E1 Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton AB, T6G 2E1 Canada
| |
Collapse
|
16
|
Origin and dynamic lineage characteristics of the developing Drosophila midgut stem cells. Dev Biol 2016; 416:347-60. [PMID: 27321560 DOI: 10.1016/j.ydbio.2016.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/01/2023]
Abstract
Proliferating intestinal stem cells (ISCs) generate all cell types of the Drosophila midgut, including enterocytes, endocrine cells, and gland cells (e.g., copper cells), throughout the lifetime of the animal. Among the signaling mechanisms controlling the balance between ISC self-renewal and the production of different cell types, Notch (N) plays a pivotal role. In this paper we investigated the emergence of ISCs during metamorphosis and the role of N in this process. Precursors of the Drosophila adult intestinal stem cells (pISCs) can be first detected within the pupal midgut during the first hours after onset of metamorphosis as motile mesenchymal cells. pISCs perform 2-3 rounds of parasynchronous divisions. The first mitosis yields only an increase in pISC number. During the following rounds of mitosis, dividing pISCs give rise to more pISCs, as well as the endocrine cells that populate the midgut of the eclosing fly. Enterocytes do not appear among the pISC progeny until around the time of eclosion. The "proendocrine" gene prospero (pros), expressed from mid-pupal stages onward in pISCs, is responsible to advance the endocrine fate in these cells; following removal of pros, pISCs continue to proliferate, but endocrine cells do not form. Conversely, the onset of N activity that occurs around the stage when pros comes on restricts pros expression among pISCs. Loss of N abrogates proliferation and switches on an endocrine fate among all pISCs. Our results suggest that a switch depending on the activity of N and pros acts at the level of the pISC to decide between continued proliferation and endocrine differentiation.
Collapse
|
17
|
Feng L, Cook B, Tsai SY, Zhou T, LaFlamme B, Evans T, Chen S. Discovery of a Small-Molecule BMP Sensitizer for Human Embryonic Stem Cell Differentiation. Cell Rep 2016; 15:2063-75. [PMID: 27210748 DOI: 10.1016/j.celrep.2016.04.066] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/06/2016] [Accepted: 04/18/2016] [Indexed: 12/19/2022] Open
Abstract
Sorely missing from the "toolkit" for directed differentiation of stem/progenitor cells are agonists of the BMP-signaling pathway. Using a high-throughput chemical screen, we discovered that PD407824, a checkpoint kinase 1 (CHK1) inhibitor, increases the sensitivity of cells to sub-threshold amounts of BMP4. We show utility of the compound in the directed differentiation of human embryonic stem cells toward mesoderm or cytotrophoblast stem cells. Blocking CHK1 activity using pharmacological compounds or CHK1 knockout using single guide RNA (sgRNA) confirmed that CHK1 inhibition increases the sensitivity to BMP4 treatment. Additional mechanistic studies indicate that CHK1 inhibition depletes p21 levels, thereby activating CDK8/9, which then phosphorylates the SMAD2/3 linker region, leading to decreased levels of SMAD2/3 protein and enhanced levels of nuclear SMAD1. This study provides insight into mechanisms controlling the BMP/transforming growth factor beta (TGF-β) signaling pathways and a useful pharmacological reagent for directed differentiation of stem cells.
Collapse
Affiliation(s)
- Lingling Feng
- Key Laboratory of Pesticide and Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, China; Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Brandoch Cook
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Su-Yi Tsai
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ting Zhou
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Brooke LaFlamme
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
18
|
Abstract
The sense of taste, or gustation, is mediated by taste buds, which are housed in specialized taste papillae found in a stereotyped pattern on the surface of the tongue. Each bud, regardless of its location, is a collection of ∼100 cells that belong to at least five different functional classes, which transduce sweet, bitter, salt, sour and umami (the taste of glutamate) signals. Taste receptor cells harbor functional similarities to neurons but, like epithelial cells, are rapidly and continuously renewed throughout adult life. Here, I review recent advances in our understanding of how the pattern of taste buds is established in embryos and discuss the cellular and molecular mechanisms governing taste cell turnover. I also highlight how these findings aid our understanding of how and why many cancer therapies result in taste dysfunction.
Collapse
Affiliation(s)
- Linda A Barlow
- Department of Cell and Developmental Biology, Graduate Program in Cell Biology, Stem Cells and Development and the Rocky Mountain Taste and Smell Center, University of Colorado, School Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
19
|
Ma M, Zhao H, Zhao H, Binari R, Perrimon N, Li Z. Wildtype adult stem cells, unlike tumor cells, are resistant to cellular damages in Drosophila. Dev Biol 2016; 411:207-216. [PMID: 26845534 DOI: 10.1016/j.ydbio.2016.01.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/31/2016] [Accepted: 01/31/2016] [Indexed: 11/26/2022]
Abstract
Adult stem cells or residential progenitor cells are critical to maintain the structure and function of adult tissues (homeostasis) throughout the lifetime of an individual. Mis-regulation of stem cell proliferation and differentiation often leads to diseases including cancer, however, how wildtype adult stem cells and cancer cells respond to cellular damages remains unclear. We find that in the adult Drosophila midgut, intestinal stem cells (ISCs), unlike tumor intestinal cells, are resistant to various cellular damages. Tumor intestinal cells, unlike wildtype ISCs, are easily eliminated by apoptosis. Further, their proliferation is inhibited upon autophagy induction, and autophagy-mediated tumor inhibition is independent of caspase-dependent apoptosis. Interestingly, inhibition of tumorigenesis by autophagy is likely through the sequestration and degradation of mitochondria, as compromising mitochondria activity in these tumor models mimics the induction of autophagy and increasing the production of mitochondria alleviates the tumor-suppression capacity of autophagy. Together, these data demonstrate that wildtype adult stem cells and tumor cells show dramatic differences in sensitivity to cellular damages, thus providing potential therapeutic implications targeting tumorigenesis.
Collapse
Affiliation(s)
- Meifang Ma
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Hang Zhao
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Hanfei Zhao
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Richard Binari
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China.
| |
Collapse
|
20
|
GATAe regulates intestinal stem cell maintenance and differentiation in Drosophila adult midgut. Dev Biol 2015; 410:24-35. [PMID: 26719127 DOI: 10.1016/j.ydbio.2015.12.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/30/2015] [Accepted: 12/19/2015] [Indexed: 12/24/2022]
Abstract
Adult intestinal tissues, exposed to the external environment, play important roles including barrier and nutrient-absorption functions. These functions are ensured by adequately controlled rapid-cell metabolism. GATA transcription factors play essential roles in the development and maintenance of adult intestinal tissues both in vertebrates and invertebrates. We investigated the roles of GATAe, the Drosophila intestinal GATA factor, in adult midgut homeostasis with its first-generated knock-out mutant as well as cell type-specific RNAi and overexpression experiments. Our results indicate that GATAe is essential for proliferation and maintenance of intestinal stem cells (ISCs). Also, GATAe is involved in the differentiation of enterocyte (EC) and enteroendocrine (ee) cells in both Notch (N)-dependent and -independent manner. The results also indicate that GATAe has pivotal roles in maintaining normal epithelial homeostasis of the Drosophila adult midgut through interaction of N signaling. Since recent reports showed that mammalian GATA-6 regulates normal and cancer stem cells in the adult intestinal tract, our data also provide information on the evolutionally conserved roles of GATA factors in stem-cell regulation.
Collapse
|
21
|
Nászai M, Carroll LR, Cordero JB. Intestinal stem cell proliferation and epithelial homeostasis in the adult Drosophila midgut. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 67:9-14. [PMID: 26024801 DOI: 10.1016/j.ibmb.2015.05.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/05/2015] [Accepted: 05/24/2015] [Indexed: 05/15/2023]
Abstract
Adult tissue homeostasis requires a tight balance between the removal of old or damaged cells and the production of new ones. Such processes are usually driven by dedicated stem cells that reside within specific tissue locations or niches. The intestinal epithelium has a remarkable regenerative capacity, which has made it a prime paradigm for the study of stem cell-driven tissue self-renewal. The discovery of the presence of stem cells in the adult midgut of the fruit fly Drosophila melanogaster has significantly impacted our understanding of the role of stem cells in intestinal homeostasis. Here we will review the current knowledge of the main mechanisms involved in the regulation of tissue homeostasis in the adult Drosophila midgut, with a focus on the role of stem cells in this process. We will also discuss processes involving acute or chronic disruption of normal intestinal homeostasis such as damage-induced regeneration and ageing.
Collapse
Affiliation(s)
- Máté Nászai
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, G61 1QH Glasgow, United Kingdom
| | - Lynsey R Carroll
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, G61 1QH Glasgow, United Kingdom
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, G61 1QH Glasgow, United Kingdom.
| |
Collapse
|
22
|
Yang J, Sun Z, Komarova NL. Analysis of stochastic stem cell models with control. Math Biosci 2015; 266:93-107. [PMID: 26073965 DOI: 10.1016/j.mbs.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 05/28/2015] [Accepted: 06/03/2015] [Indexed: 12/11/2022]
Abstract
Understanding the dynamics of stem cell lineages is of central importance both for healthy and cancerous tissues. We study stochastic population dynamics of stem cells and differentiated cells, where cell decisions, such as proliferation vs. differentiation decisions, or division and death decisions, are under regulation from surrounding cells. The goal is to understand how different types of control mechanisms affect the means and variances of cell numbers. We use the assumption of weak dependencies of the regulatory functions (the controls) on the cell populations near the equilibrium to formulate moment equations. We then study three different methods of closure, showing that they all lead to the same results for the highest order terms in the expressions for the moments. We derive simple explicit expressions for the means and the variances of stem cell and differentiated cell numbers. It turns out that the variance is expressed as an algebraic function of partial derivatives of the controls with respect to the population sizes at the equilibrium. We demonstrate that these findings are consistent with the results previously obtained in the context of particular systems, and also present two novel examples with negative and positive control of division and differentiation decisions. This methodology is formulated without any specific assumptions on the functional form of the controls, and thus can be used for any biological system.
Collapse
Affiliation(s)
- Jienian Yang
- Department of Mathematics, University of California Irvine, Irvine, CA 92617, United States
| | - Zheng Sun
- Department of Mathematics, University of California Irvine, Irvine, CA 92617, United States
| | - Natalia L Komarova
- Department of Mathematics, University of California Irvine, Irvine, CA 92617, United States.
| |
Collapse
|
23
|
Wołczuk K, Nowakowska J, Płąchocki D, Kakareko T. Histological, histochemical and ultrastructural analysis reveals functional division of the oesophagogastric segment in freshwater tubenose goby Proterorhinus semilunaris Heckel, 1837. ZOOMORPHOLOGY 2014; 134:259-268. [PMID: 25995536 PMCID: PMC4430593 DOI: 10.1007/s00435-014-0250-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 11/15/2014] [Accepted: 11/18/2014] [Indexed: 01/20/2023]
Abstract
Histological and histochemical features of the oesophagogastric segment of the alimentary canal as well as ultrastructure of gastric gland cells of freshwater tubenose goby Proterorhinus semilunaris were examined. The studies revealed that despite the lack of anatomical distinction, the oesophagogastric segment is histologically divided into the oesophagus, oesogaster and stomach, which provides evidence for the functional compartmentation of this organ. The oesophagus was characterised by the presence of numerous goblet cells secreting mainly a mixture of neutral and acid mucopolysaccharides. In the stomach, the apical zone of the surface epithelial cells contained neutral mucopolysaccharides. Numerous proliferating cells were scattered throughout the surface epithelium. In the lamina propria of the stomach, a well-developed layer of gastric glands was observed. The glands were of the alveolar type and occupied nearly the entire length of the stomach except the pyloric region. The gastric gland cells were varied into light and dark; however, their ultrastructure was identical. All cells had numerous mitochondria and a well-developed tubulovesicular system typical for the oxynticopeptic cells, but pepsinogen granules were not present in the cytoplasm of these cells. These findings contribute new evidence to literature reports that not all gobiid fish are stomachless. Moreover, they suggest higher adaptation of the species to utilise protein-rich food compared to stomachless fish, and its ability to adjust the alimentary canal quickly to changing diet. How this may facilitate establishment of P. semilunaris in invaded environments remains an open question.
Collapse
Affiliation(s)
- Katarzyna Wołczuk
- Department of Zoology of Vertebrates, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
| | - Julita Nowakowska
- Laboratory of Electron and Confocal Microscopy, University of Warsaw, Miecznikowa 1, Warsaw, Poland
| | - Dariusz Płąchocki
- Department of Hydrobiology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
| | - Tomasz Kakareko
- Department of Hydrobiology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
| |
Collapse
|
24
|
Petkau K, Parsons BD, Duggal A, Foley E. A deregulated intestinal cell cycle program disrupts tissue homeostasis without affecting longevity in Drosophila. J Biol Chem 2014; 289:28719-29. [PMID: 25170078 DOI: 10.1074/jbc.m114.578708] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Recent studies illuminate a complex relationship between the control of stem cell division and intestinal tissue organization in the model system Drosophila melanogaster. Host and microbial signals drive intestinal proliferation to maintain an effective epithelial barrier. Although it is widely assumed that proliferation induces dysplasia and shortens the life span of the host, the phenotypic consequences of deregulated intestinal proliferation for an otherwise healthy host remain unexplored. To address this question, we genetically isolated and manipulated the cell cycle programs of adult stem cells and enterocytes. Our studies revealed that cell cycle alterations led to extensive cell death and morphological disruptions. Despite the extensive tissue damage, we did not observe an impact on longevity, suggesting a remarkable degree of plasticity in intestinal function.
Collapse
Affiliation(s)
- Kristina Petkau
- From the Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Brendon D Parsons
- From the Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Aashna Duggal
- From the Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Edan Foley
- From the Department of Medical Microbiology and Immunology, Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
25
|
Abstract
The digestive tract plays a central role in the digestion and absorption of nutrients. Far from being a passive tube, it provides the first line of defense against pathogens and maintains energy homeostasis by exchanging neuronal and endocrine signals with other organs. Historically neglected, the gut of the fruit fly Drosophila melanogaster has recently come to the forefront of Drosophila research. Areas as diverse as stem cell biology, neurobiology, metabolism, and immunity are benefitting from the ability to study the genetics of development, growth regulation, and physiology in the same organ. In this review, we summarize our knowledge of the Drosophila digestive tract, with an emphasis on the adult midgut and its functional underpinnings.
Collapse
Affiliation(s)
- Bruno Lemaitre
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale Lausanne (EPFL), CH-1015 Lausanne, Switzerland;
| | | |
Collapse
|
26
|
Modeling colorectal cancer as a 3-dimensional disease in a dish: the case for drug screening using organoids, zebrafish, and fruit flies. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 10:e73-81. [PMID: 24050233 DOI: 10.1016/j.ddtec.2012.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This review discusses recent shifts in the understanding of colorectal cancer as a stem cell based disease, based on findings that tie patient prognosis to the presence of cancer stem cells in colorectal tumors. Currently no drugs specifically target CSCs in colorectal tumors. However, recent advances in the culturing of colorectal stem cells using mammalian organoids, zebrafish, and Drosophila offer promising avenues for anti-CSC drug discovery.
Collapse
|
27
|
Abstract
Here we report the development of an in vivo system to study the interaction of stem cells with drugs using a tumor model in the adult Drosophila intestine. Strikingly, we find that some Food and Drug Administration-approved chemotherapeutics that can inhibit the growth of Drosophila tumor stem cells can paradoxically promote the hyperproliferation of their wild-type counterparts. These results reveal an unanticipated side effect on stem cells that may contribute to tumor recurrence. We propose that the same side effect may occur in humans based on our finding that it is driven in Drosophila by the evolutionarily conserved Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway. An immediate implication of our findings is that supplementing traditional chemotherapeutics with anti-inflammatories may reduce tumor recurrence.
Collapse
|
28
|
Wang C, Guo X, Xi R. EGFR and Notch signaling respectively regulate proliferative activity and multiple cell lineage differentiation of Drosophila gastric stem cells. Cell Res 2014; 24:610-27. [PMID: 24603358 DOI: 10.1038/cr.2014.27] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 12/16/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022] Open
Abstract
Quiescent, multipotent gastric stem cells (GSSCs) in the copper cell region of adult Drosophila midgut can produce all epithelial cell lineages found in the region, including acid-secreting copper cells, interstitial cells and enteroendocrine cells, but mechanisms controlling their quiescence and the ternary lineage differentiation are unknown. By using cell ablation or damage-induced regeneration assays combined with cell lineage tracing and genetic analysis, here we demonstrate that Delta (Dl)-expressing cells in the copper cell region are the authentic GSSCs that can self-renew and continuously regenerate the gastric epithelium after a sustained damage. Lineage tracing analysis reveals that the committed GSSC daughter with activated Notch will invariably differentiate into either a copper cell or an interstitial cell, but not the enteroendocrine cell lineage, and loss-of-function and gain-of-function studies revealed that Notch signaling is both necessary and sufficient for copper cell/interstitial cell differentiation. We also demonstrate that elevated epidermal growth factor receptor (EGFR) signaling, which is achieved by the activation of ligand Vein from the surrounding muscle cells and ligand Spitz from progenitor cells, mediates the regenerative proliferation of GSSCs following damage. Taken together, we demonstrate that Dl is a specific marker for Drosophila GSSCs, whose cell cycle status is dependent on the levels of EGFR signaling activity, and the Notch signaling has a central role in controlling cell lineage differentiation from GSSCs by separating copper/interstitial cell lineage from enteroendocrine cell lineage.
Collapse
Affiliation(s)
- Chenhui Wang
- 1] National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China [2] College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xingting Guo
- 1] National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China [2] College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Rongwen Xi
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| |
Collapse
|
29
|
Okumura T, Takeda K, Taniguchi K, Adachi-Yamada T. βν integrin inhibits chronic and high level activation of JNK to repress senescence phenotypes in Drosophila adult midgut. PLoS One 2014; 9:e89387. [PMID: 24586740 PMCID: PMC3930726 DOI: 10.1371/journal.pone.0089387] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 01/20/2014] [Indexed: 01/24/2023] Open
Abstract
Proper control of adult stem cells including their proliferation and differentiation is crucial in maintaining homeostasis of well-organized tissues/organs throughout an organism's life. The Drosophila adult midgut has intestinal stem cells (ISCs), which have been exploited as a simple model system to investigate mechanisms controlling adult tissue homeostasis. Here, we found that a viable mutant of βν integrin (βint-ν), encoding one of two Drosophila integrin β subunits, showed a short midgut and abnormal multilayered epithelia accompanied by an increase in ISC proliferation and misdifferentiation defects. The increase in ISC proliferation and misdifferentiation was due to frequent ISC duplication expanding a pool of ISCs, which was caused by depression of the Notch signalling, and up-regulation of unpaired (upd), a gene encoding an extracellular ligand in the JAK/STAT signalling pathway. In addition, we observed that abnormally high accumulation of filamentous actin (F-actin) was caused in the βint-ν mutant enterocytes. Furthermore, the defects were rescued by suppressing c-Jun N-terminal kinase (JNK) signalling, which was up-regulated in a manner correlated with the defect levels in the above-mentioned βint-ν mutant phenotype. These symptoms observed in young βint-ν mutant midgut were very similar to those in the aged midgut in wild type. Our results suggested that βint-ν has a novel function for the Drosophila adult midgut homeostasis under normal conditions and provided a new insight into possible age-related diseases caused by latent abnormality of an integrin function.
Collapse
Affiliation(s)
- Takashi Okumura
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Koji Takeda
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
- Graduate Course in Life Science, Graduate School of Science, Gakushuin University, Tokyo, Japan
| | - Kiichiro Taniguchi
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Takashi Adachi-Yamada
- Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
- Graduate Course in Life Science, Graduate School of Science, Gakushuin University, Tokyo, Japan
- Institute for Biomolecular Science, Gakushuin University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
30
|
Shim J, Gururaja-Rao S, Banerjee U. Nutritional regulation of stem and progenitor cells in Drosophila. Development 2014; 140:4647-56. [PMID: 24255094 DOI: 10.1242/dev.079087] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stem cells and their progenitors are maintained within a microenvironment, termed the niche, through local cell-cell communication. Systemic signals originating outside the niche also affect stem cell and progenitor behavior. This review summarizes studies that pertain to nutritional effects on stem and progenitor cell maintenance and proliferation in Drosophila. Multiple tissue types are discussed that utilize the insulin-related signaling pathway to convey nutritional information either directly to these progenitors or via other cell types within the niche. The concept of systemic control of these cell types is not limited to Drosophila and may be functional in vertebrate systems, including mammals.
Collapse
Affiliation(s)
- Jiwon Shim
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
31
|
Abstract
Identifying the exact regulatory circuits that can stably maintain tissue homeostasis is critical for our basic understanding of multicellular organisms, and equally critical for identifying how tumors circumvent this regulation, thus providing targets for treatment. Despite great strides in the understanding of the molecular components of stem-cell regulation, the overall mechanisms orchestrating tissue homeostasis are still far from being understood. Typically, tissue contains the stem cells, transit amplifying cells, and terminally differentiated cells. Each of these cell types can potentially secrete regulatory factors and/or respond to factors secreted by other types. The feedback can be positive or negative in nature. This gives rise to a bewildering array of possible mechanisms that drive tissue regulation. In this paper, we propose a novel method of studying stem cell lineage regulation, and identify possible numbers, types, and directions of control loops that are compatible with stability, keep the variance low, and possess a certain degree of robustness. For example, there are exactly two minimal (two-loop) control networks that can regulate two-compartment (stem and differentiated cell) tissues, and 20 such networks in three-compartment tissues. If division and differentiation decisions are coupled, then there must be a negative control loop regulating divisions of stem cells (e.g. by means of contact inhibition). While this mechanism is associated with the highest robustness, there could be systems that maintain stability by means of positive divisions control, coupled with specific types of differentiation control. Some of the control mechanisms that we find have been proposed before, but most of them are new, and we describe evidence for their existence in data that have been previously published. By specifying the types of feedback interactions that can maintain homeostasis, our mathematical analysis can be used as a guide to experimentally zero in on the exact molecular mechanisms in specific tissues.
Collapse
Affiliation(s)
- Natalia L. Komarova
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
32
|
Li L, Wang J, Chau JFL, Liu H, Li B, Hao A, Li J. Smad1 stabilization and delocalization in response to the blockade of BMP activity. Cell Mol Biol Lett 2013; 18:340-54. [PMID: 23793844 PMCID: PMC6275640 DOI: 10.2478/s11658-013-0093-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 06/13/2013] [Indexed: 01/22/2023] Open
Abstract
Signaling at the plasma membrane receptors is generally terminated by some form of feedback regulation, such as endocytosis and/or degradation of the receptors. BMP-Smad1 signaling can also be attenuated by BMP-induced expression of the inhibitory Smads, which are negative regulators of Smad1 transactivation activity and/or BMP antagonists. Here, we report on a novel Smad1 regulation mechanism that occurs in response to the blockade of BMP activity. Lowering the serum levels or antagonizing BMPs with noggin led to upregulation of Smad1 at the protein level in several cell lines, but not to upregulation of Smad5, Smad8 or Smad2/3. The Smad1 upregulation occurs at the level of protein stabilization. Upregulated Smad1 was relocalized to the perinuclear region. These alterations seem to affect the dynamics and amplitude of BMP2-induced Smad1 reactivation. Our findings indicate that depleting or antagonizing BMPs leads to Smad1 stabilization and relocalization, thus revealing an unexpected regulatory mechanism for BMP-Smad1 signaling.
Collapse
Affiliation(s)
- Lili Li
- Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong, 250012 China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhe Wang
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | | | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Aijun Hao
- Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong, 250012 China
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Panayidou S, Apidianakis Y. Regenerative inflammation: lessons from Drosophila intestinal epithelium in health and disease. Pathogens 2013; 2:209-31. [PMID: 25437036 PMCID: PMC4235722 DOI: 10.3390/pathogens2020209] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/18/2013] [Accepted: 03/22/2013] [Indexed: 01/04/2023] Open
Abstract
Intestinal inflammation is widely recognized as a pivotal player in health and disease. Defined cytologically as the infiltration of leukocytes in the lamina propria layer of the intestine, it can damage the epithelium and, on a chronic basis, induce inflammatory bowel disease and potentially cancer. The current view thus dictates that blood cell infiltration is the instigator of intestinal inflammation and tumor-promoting inflammation. This is based partially on work in humans and mice showing that intestinal damage during microbially mediated inflammation activates phagocytic cells and lymphocytes that secrete inflammatory signals promoting tissue damage and tumorigenesis. Nevertheless, extensive parallel work in the Drosophila midgut shows that intestinal epithelium damage induces inflammatory signals and growth factors acting mainly in a paracrine manner to induce intestinal stem cell proliferation and tumor formation when genetically predisposed. This is accomplished without any apparent need to involve Drosophila hemocytes. Therefore, recent work on Drosophila host defense to infection by expanding its main focus on systemic immunity signaling pathways to include the study of organ homeostasis in health and disease shapes a new notion that epithelially emanating cytokines and growth factors can directly act on the intestinal stem cell niche to promote “regenerative inflammation” and potentially cancer.
Collapse
Affiliation(s)
- Stavria Panayidou
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus.
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus.
| |
Collapse
|
34
|
The Fruit Fly Drosophila melanogaster as a Model for Aging Research. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2013; 135:63-77. [DOI: 10.1007/10_2013_193] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
35
|
Stem cells in the context of evolution and development. Dev Genes Evol 2012; 223:1-3. [PMID: 23223955 DOI: 10.1007/s00427-012-0430-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
|
36
|
|