1
|
Sebo DJ, Ali I, Fetsko AR, Trimbach AA, Taylor MR. Activation of Wnt/β-catenin in neural progenitor cells regulates blood-brain barrier development and promotes neuroinflammation. Sci Rep 2025; 15:3496. [PMID: 39875426 PMCID: PMC11775206 DOI: 10.1038/s41598-025-85784-8] [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: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The central nervous system (CNS) requires specialized blood vessels to support neural function within specific microenvironments. During neurovascular development, endothelial Wnt/β-catenin signaling is required for BBB development within the brain parenchyma, whereas fenestrated blood vessels that lack BBB properties do not require Wnt/β-catenin signaling. Here, we used zebrafish to further characterize this phenotypic heterogeneity of the CNS vasculature. Using transgenic reporters of Wnt/β-catenin transcriptional activity, we found an inverse correlation between activated Wnt/β-catenin signaling in endothelial cells (ECs) versus non-ECs within these distinct microenvironments. Our results indicated that the level of Wnt/β-catenin signaling in non-ECs may regulate Wnt/β-catenin activity in adjacent ECs. To further test this concept, we generated a transgenic Tet-On inducible system to drive constitutively active β-catenin expression in neural progenitor cells (NPCs). We found that dose-dependent activation of Wnt/β-catenin in NPCs caused severe deficiency in CNS angiogenesis and BBB development. Additionally, we discovered a significant increase in the proliferation of microglia and infiltration of peripheral neutrophils indicative of a stereotypical neuroinflammatory response. In conclusion, our results demonstrate the importance of proper Wnt/β-catenin signaling within specific CNS microenvironments and highlights the potentially deleterious consequences of aberrant Wnt activation.
Collapse
Affiliation(s)
- Dylan J Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Irshad Ali
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Audrey R Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Aubrey A Trimbach
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael R Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Yu T, Chen J, Wang Y, Xu J. The embryonic zebrafish brain is exclusively colonized by pu.1-dependent and lymphatic-independent population of microglia. SCIENCE ADVANCES 2024; 10:eado0519. [PMID: 39196933 PMCID: PMC11352844 DOI: 10.1126/sciadv.ado0519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/24/2024] [Indexed: 08/30/2024]
Abstract
Microglia, the crucial immune cells inhabiting the central nervous system (CNS), perform a range of vital functions, encompassing immune defense and neuronal regulation. Microglia subsets with diverse functions and distinct developmental regulations have been identified recently. It is generally accepted that all microglia originate from hematopoiesis and depend on the myeloid transcription factor PU.1. However, a recent study reported the existence of mrc1+ microglia in zebrafish embryos, which are seemingly independent of Pu.1 and reliant on lymphatic vessels, sparking great interest in the possibility of lymphatic-originated microglia. To address this, we took advantage of a pu.1 knock-in zebrafish allele for a detailed investigation. Our results conclusively showed that almost all zebrafish embryonic microglia (~95% on average) express pu.1. Further, lineage tracing and mutant analysis revealed that these microglia neither emerged from nor depended on lymphatic vessels. In essence, our study refutes the presence of pu.1-independent but lymphatic-dependent microglia.
Collapse
Affiliation(s)
- Tao Yu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Jiahao Chen
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Yuexin Wang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Jin Xu
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
3
|
Arroyo AB, Tyrkalska SD, Bastida-Martínez E, Monera-Girona AJ, Cantón-Sandoval J, Bernal-Carrión M, García-Moreno D, Elías-Arnanz M, Mulero V. Peds1 deficiency in zebrafish results in myeloid cell apoptosis and exacerbated inflammation. Cell Death Discov 2024; 10:388. [PMID: 39209813 PMCID: PMC11362147 DOI: 10.1038/s41420-024-02141-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Plasmalogens are glycerophospholipids with a vinyl ether bond that confers unique properties. Recent identification of the gene encoding PEDS1, the desaturase generating the vinyl ether bond, enables evaluation of the role of plasmalogens in health and disease. Here, we report that Peds1-deficient zebrafish larvae display delayed development, increased basal inflammation, normal hematopoietic stem and progenitor cell emergence, and cell-autonomous myeloid cell apoptosis. In a sterile acute inflammation model, Peds1-deficient larvae exhibited impaired inflammation resolution and tissue regeneration, increased interleukin-1β and NF-κB expression, and elevated ROS levels at the wound site. Abnormal immune cell recruitment, neutrophil persistence, and fewer but predominantly pro-inflammatory macrophages were observed. Chronic skin inflammation worsened in Peds1-deficient larvae but was mitigated by exogenous plasmalogen, which also alleviated hyper-susceptibility to bacterial infection, as did pharmacological inhibition of caspase-3 and colony-stimulating factor 3-induced myelopoiesis. Overall, our results highlight an important role for plasmalogens in myeloid cell biology and inflammation.
Collapse
Affiliation(s)
- Ana B Arroyo
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Sylwia D Tyrkalska
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Eva Bastida-Martínez
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
| | - Antonio J Monera-Girona
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
| | - Joaquín Cantón-Sandoval
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Martín Bernal-Carrión
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
| | - Diana García-Moreno
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Montserrat Elías-Arnanz
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
| | - Victoriano Mulero
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
4
|
Yamamoto J, Deguchi H, Sumiyoshi T, Nakagami K, Saito A, Miyanishi H, Kondo M, Kono T, Sakai M, Kinoshita M, Hikima JI. Accumulation and Phagocytosis of Fluorescently Visualized Macrophages Against Edwardsiella piscicida Infection in Established mpeg1.1-Transgenic Japanese Medaka Oryzias latipes. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:658-671. [PMID: 38888725 DOI: 10.1007/s10126-024-10333-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Intracellular bacteria such as those belonging to the genus Edwardsiella can survive and proliferate within macrophages. However, the detailed mechanisms underlying the host macrophage immune response and pathogen evasion strategies remain unknown. To advance the field of host macrophage research, we successfully established transgenic (Tg) Japanese medaka Oryzias latipes that possesses fluorescently visualized macrophages. As a macrophage marker, the macrophage-expressed gene 1.1 (mpeg1.1) was selected because of its predominant expression across various tissues in medaka. To validate the macrophage characteristics of the fluorescently labeled cells, May-Grünwald Giemsa staining and peroxidase staining were conducted. The labeled cells exhibited morphological features consistent with those of monocyte/macrophage-like cells and tested negative for peroxidase activity. Through co-localization studies, the fluorescently labeled cells co-localized with E. piscicida in the intestines and kidneys of infected medaka larvae, confirming the ingestion of bacteria through phagocytosis. In addition, the labeled cells expressed macrophage markers but lacked a neutrophil marker. These results suggested that the fluorescently labeled cells of Tg[mpeg1.1:mCherry/mAG] medaka were monocytes/macrophages, which will be useful for future studies aimed at understanding the mechanisms of macrophage-mediated bacterial infections.
Collapse
Affiliation(s)
- Juna Yamamoto
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Hana Deguchi
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Takechiyo Sumiyoshi
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Kentaro Nakagami
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Akatsuki Saito
- Department of Veterinary Medicine, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Hiroshi Miyanishi
- Department of Marine Biology and Environmental Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Masakazu Kondo
- Department of Applied Aquabiology, National Fisheries University, Japan Fisheries Research and Education Agency, Yamaguchi, 759-6595, Japan
| | - Tomoya Kono
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Masahiro Sakai
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Masato Kinoshita
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Jun-Ichi Hikima
- Course of Biochemistry and Applied Biosciences, Graduate School of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
| |
Collapse
|
5
|
Zhao C, Yang Z, Li Y, Wen Z. Macrophages in tissue repair and regeneration: insights from zebrafish. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:12. [PMID: 38861103 PMCID: PMC11166613 DOI: 10.1186/s13619-024-00195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Macrophages play crucial and versatile roles in regulating tissue repair and regeneration upon injury. However, due to their complex compositional heterogeneity and functional plasticity, deciphering the nature of different macrophage subpopulations and unraveling their dynamics and precise roles during the repair process have been challenging. With its distinct advantages, zebrafish (Danio rerio) has emerged as an invaluable model for studying macrophage development and functions, especially in tissue repair and regeneration, providing valuable insights into our understanding of macrophage biology in health and diseases. In this review, we present the current knowledge and challenges associated with the role of macrophages in tissue repair and regeneration, highlighting the significant contributions made by zebrafish studies. We discuss the unique advantages of the zebrafish model, including its genetic tools, imaging techniques, and regenerative capacities, which have greatly facilitated the investigation of macrophages in these processes. Additionally, we outline the potential of zebrafish research in addressing the remaining challenges and advancing our understanding of the intricate interplay between macrophages and tissue repair and regeneration.
Collapse
Affiliation(s)
- Changlong Zhao
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiyong Yang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yunbo Li
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zilong Wen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Habjan E, Schouten GK, Speer A, van Ulsen P, Bitter W. Diving into drug-screening: zebrafish embryos as an in vivo platform for antimicrobial drug discovery and assessment. FEMS Microbiol Rev 2024; 48:fuae011. [PMID: 38684467 PMCID: PMC11078164 DOI: 10.1093/femsre/fuae011] [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: 11/01/2023] [Revised: 02/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
The rise of multidrug-resistant bacteria underlines the need for innovative treatments, yet the introduction of new drugs has stagnated despite numerous antimicrobial discoveries. A major hurdle is a poor correlation between promising in vitro data and in vivo efficacy in animal models, which is essential for clinical development. Early in vivo testing is hindered by the expense and complexity of existing animal models. Therefore, there is a pressing need for cost-effective, rapid preclinical models with high translational value. To overcome these challenges, zebrafish embryos have emerged as an attractive model for infectious disease studies, offering advantages such as ethical alignment, rapid development, ease of maintenance, and genetic manipulability. The zebrafish embryo infection model, involving microinjection or immersion of pathogens and potential antibiotic hit compounds, provides a promising solution for early-stage drug screening. It offers a cost-effective and rapid means of assessing the efficacy, toxicity and mechanism of action of compounds in a whole-organism context. This review discusses the experimental design of this model, but also its benefits and challenges. Additionally, it highlights recently identified compounds in the zebrafish embryo infection model and discusses the relevance of the model in predicting the compound's clinical potential.
Collapse
Affiliation(s)
- Eva Habjan
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Gina K Schouten
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Alexander Speer
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Peter van Ulsen
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
7
|
Speirs ZC, Loynes CA, Mathiessen H, Elks PM, Renshaw SA, Jørgensen LVG. What can we learn about fish neutrophil and macrophage response to immune challenge from studies in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109490. [PMID: 38471626 DOI: 10.1016/j.fsi.2024.109490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Fish rely, to a high degree, on the innate immune system to protect them against the constant exposure to potential pathogenic invasion from the surrounding water during homeostasis and injury. Zebrafish larvae have emerged as an outstanding model organism for immunity. The cellular component of zebrafish innate immunity is similar to the mammalian innate immune system and has a high degree of sophistication due to the needs of living in an aquatic environment from early embryonic stages of life. Innate immune cells (leukocytes), including neutrophils and macrophages, have major roles in protecting zebrafish against pathogens, as well as being essential for proper wound healing and regeneration. Zebrafish larvae are visually transparent, with unprecedented in vivo microscopy opportunities that, in combination with transgenic immune reporter lines, have permitted visualisation of the functions of these cells when zebrafish are exposed to bacterial, viral and parasitic infections, as well as during injury and healing. Recent findings indicate that leukocytes are even more complex than previously anticipated and are essential for inflammation, infection control, and subsequent wound healing and regeneration.
Collapse
Affiliation(s)
- Zoë C Speirs
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Catherine A Loynes
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Heidi Mathiessen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Stephen A Renshaw
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Louise von Gersdorff Jørgensen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark.
| |
Collapse
|
8
|
Wright K, Han DJ, Song R, de Silva K, Plain KM, Purdie AC, Shepherd A, Chin M, Hortle E, Wong JJL, Britton WJ, Oehlers SH. Zebrafish tsc1 and cxcl12a increase susceptibility to mycobacterial infection. Life Sci Alliance 2024; 7:e202302523. [PMID: 38307625 PMCID: PMC10837051 DOI: 10.26508/lsa.202302523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024] Open
Abstract
Regulation of host miRNA expression is a contested node that controls the host immune response to mycobacterial infection. The host must counter subversive efforts of pathogenic mycobacteria to launch a protective immune response. Here, we examine the role of miR-126 in the zebrafish-Mycobacterium marinum infection model and identify a protective role for infection-induced miR-126 through multiple effector pathways. We identified a putative link between miR-126 and the tsc1a and cxcl12a/ccl2/ccr2 signalling axes resulting in the suppression of non-tnfa expressing macrophage accumulation at early M. marinum granulomas. Mechanistically, we found a detrimental effect of tsc1a expression that renders zebrafish embryos susceptible to higher bacterial burden and increased cell death via mTOR inhibition. We found that macrophage recruitment driven by the cxcl12a/ccl2/ccr2 signalling axis was at the expense of the recruitment of classically activated tnfa-expressing macrophages and increased cell death around granulomas. Together, our results delineate putative pathways by which infection-induced miR-126 may shape an effective immune response to M. marinum infection in zebrafish embryos.
Collapse
Affiliation(s)
- Kathryn Wright
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia
- Directed Evolution Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Darryl Jy Han
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Renhua Song
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Epigenetics and RNA Biology Laboratory, Charles Perkins Centre, The University of Sydney, Camperdown, Australia
| | - Kumudika de Silva
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia
| | - Karren M Plain
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia
| | - Auriol C Purdie
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia
| | - Ava Shepherd
- Directed Evolution Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
| | - Maegan Chin
- Directed Evolution Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
| | - Elinor Hortle
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Faculty of Science, School of Life Sciences, Centre for Inflammation and University of Technology Sydney, Sydney, Australia
| | - Justin J-L Wong
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Epigenetics and RNA Biology Laboratory, Charles Perkins Centre, The University of Sydney, Camperdown, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
9
|
Tanner CD, Rosowski EE. Macrophages inhibit extracellular hyphal growth of A. fumigatus through Rac2 GTPase signaling. Infect Immun 2024; 92:e0038023. [PMID: 38168666 PMCID: PMC10863406 DOI: 10.1128/iai.00380-23] [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: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Macrophages act as a first line of defense against pathogens. Against Aspergillus fumigatus, a fungus with pathogenic potential in immunocompromised patients, macrophages can phagocytose fungal spores and inhibit spore germination to prevent the development of tissue-invasive hyphae. However, the cellular pathways that macrophages use to accomplish these tasks and any roles macrophages have later in infection against invasive forms of fungi are still not fully known. Rac-family Rho GTPases are signaling hubs for multiple cellular functions in leukocytes, including cell migration, phagocytosis, reactive oxygen species (ROS) generation, and transcriptional activation. We therefore aimed to further characterize the function of macrophages against A. fumigatus in an in vivo vertebrate infection model by live imaging of the macrophage behavior in A. fumigatus-infected rac2 mutant zebrafish larvae. While Rac2-deficient zebrafish larvae are susceptible to A. fumigatus infection, Rac2 deficiency does not impair macrophage migration to the infection site, interaction with and phagocytosis of spores, spore trafficking to acidified compartments, or spore killing. However, we reveal a role for Rac2 in macrophage-mediated inhibition of spore germination and control of invasive hyphae. Re-expression of Rac2 under a macrophage-specific promoter rescues the survival of A. fumigatus-infected rac2 mutant larvae through increased control of germination and hyphal growth. Altogether, we describe a new role for macrophages against extracellular hyphal growth of A. fumigatus and report that the function of the Rac2 Rho GTPase in macrophages is required for this function.
Collapse
Affiliation(s)
- Christopher D. Tanner
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Emily E. Rosowski
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
10
|
Shin K, Begeman IJ, Cao J, Kang J. leptin b and its regeneration enhancer illustrate the regenerative features of zebrafish hearts. Dev Dyn 2024; 253:91-106. [PMID: 36495292 PMCID: PMC10256838 DOI: 10.1002/dvdy.556] [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: 07/14/2022] [Revised: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Zebrafish possess a remarkable regenerative capacity, which is mediated by the induction of various genes upon injury. Injury-dependent transcription is governed by the tissue regeneration enhancer elements (TREEs). Here, we utilized leptin b (lepb), an injury-specific factor, and its TREE to dissect heterogeneity of noncardiomyocytes (CMs) in regenerating hearts. RESULTS Our single-cell RNA sequencing (scRNA-seq) analysis demonstrated that the endothelium/endocardium(EC) is activated to induce distinct subpopulations upon injury. We demonstrated that lepb can be utilized as a regeneration-specific marker to subset injury-activated ECs. lepb+ ECs robustly induce pro-regenerative factors, implicating lepb+ ECs as a signaling center to interact with other cardiac cells. Our scRNA-seq analysis identified that lepb is also produced by subpopulation of epicardium (Epi) and epicardium-derived cells (EPDCs). To determine whether lepb labels injury-emerging non-CM cells, we tested the activity of lepb-linked regeneration enhancer (LEN) with chromatin accessibility profiles and transgenic lines. While nondetectable in uninjured hearts, LEN directs EC and Epi/EPDC expression upon injury. The endogenous LEN activity was assessed using LEN deletion lines, demonstrating that LEN deletion abolished injury-dependent expression of lepb, but not other nearby genes. CONCLUSIONS Our integrative analyses identify regeneration-emerging cell-types and factors, leading to the discovery of regenerative features of hearts.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Ian J. Begeman
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10021, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| |
Collapse
|
11
|
Michael C, de Oliveira S. Exploring the dynamic behavior of leukocytes with zebrafish. Curr Opin Cell Biol 2023; 85:102276. [PMID: 37956533 PMCID: PMC10842401 DOI: 10.1016/j.ceb.2023.102276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023]
Abstract
Cell migration is a complex and intricate network of physical, chemical, and molecular events that ultimately leads to cell motility. This phenomenon is involved in both physiological and pathological processes such as proper immune and inflammatory responses. Dysregulation of cell migration machinery in immune cells can have a tremendous impact on the trajectory of inflammation, infection, and resolution. The small vertebrate, the zebrafish, has a remarkable capacity for genetic and pharmacological manipulation aligned to transparency that enables modulation and visualization of cell migration in vivo noninvasively. Such characteristics revolutionized the field of leukocyte biology, particularly neutrophils. In this review, we will focus on leukocyte migration and highlight findings made in the zebrafish that demonstrate how this small vertebrate system is a unique model to perform in vivo imaging and study mechanisms that regulate the dynamic behavior of immune cells in their native environment under homeostasis or upon challenge.
Collapse
Affiliation(s)
- Cassia Michael
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Montefiore-Einstein Comprehensive Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Cancer Dormancy Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, USA.
| |
Collapse
|
12
|
Palsamy K, Chen JY, Skaggs K, Qadeer Y, Connors M, Cutler N, Richmond J, Kommidi V, Poles A, Affrunti D, Powell C, Goldman D, Parent JM. Microglial depletion after brain injury prolongs inflammation and impairs brain repair, adult neurogenesis and pro-regenerative signaling. Glia 2023; 71:2642-2663. [PMID: 37449457 PMCID: PMC10528132 DOI: 10.1002/glia.24444] [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: 02/16/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The adult zebrafish brain, unlike mammals, has a remarkable regenerative capacity. Although inflammation in part hinders regeneration in mammals, it is necessary for zebrafish brain repair. Microglia are resident brain immune cells that regulate the inflammatory response. To explore the microglial role in repair, we used liposomal clodronate or colony stimulating factor-1 receptor (csf1r) inhibitor to suppress microglia after brain injury, and also examined regeneration in two genetic mutant lines that lack microglia. We found that microglial ablation impaired telencephalic regeneration after injury. Microglial suppression attenuated cell proliferation at the intermediate progenitor cell amplification stage of neurogenesis. Notably, the loss of microglia impaired phospho-Stat3 (signal transducer and activator of transcription 3) and ß-Catenin signaling after injury. Furthermore, the ectopic activation of Stat3 and ß-Catenin rescued neurogenesis defects caused by microglial loss. Microglial suppression also prolonged the post-injury inflammatory phase characterized by neutrophil accumulation, likely hindering the resolution of inflammation. These findings reveal specific roles of microglia and inflammatory signaling during zebrafish telencephalic regeneration that should advance strategies to improve mammalian brain repair.
Collapse
Affiliation(s)
- Kanagaraj Palsamy
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica Y Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kaia Skaggs
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- University of Findlay, Findlay, Ohio, USA
| | - Yusuf Qadeer
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Meghan Connors
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Cutler
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joshua Richmond
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Vineeth Kommidi
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison Poles
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Danielle Affrunti
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Curtis Powell
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Goldman
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Rodríguez‐Ruiz L, Lozano‐Gil JM, Naranjo‐Sánchez E, Martínez‐Balsalobre E, Martínez‐López A, Lachaud C, Blanquer M, Phung TK, García‐Moreno D, Cayuela ML, Tyrkalska SD, Pérez‐Oliva AB, Mulero V. ZAKα/P38 kinase signaling pathway regulates hematopoiesis by activating the NLRP1 inflammasome. EMBO Mol Med 2023; 15:e18142. [PMID: 37675820 PMCID: PMC10565642 DOI: 10.15252/emmm.202318142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Chronic inflammatory diseases are associated with hematopoietic lineage bias, including neutrophilia and anemia. We have recently identified that the canonical inflammasome mediates the cleavage of the master erythroid transcription factor GATA1 in hematopoietic stem and progenitor cells (HSPCs). We report here that genetic inhibition of Nlrp1 resulted in reduced number of neutrophils and increased erythrocyte counts in zebrafish larvae. We also found that the NLRP1 inflammasome in human cells was inhibited by LRRFIP1 and FLII, independently of DPP9, and both inhibitors regulated hematopoiesis. Mechanistically, erythroid differentiation resulted in ribosomal stress-induced activation of the ZAKα/P38 kinase axis which, in turn, phosphorylated and promoted the assembly of NLRP1 in both zebrafish and human. Finally, inhibition of Zaka with the FDA/EMA-approved drug Nilotinib alleviated neutrophilia in a zebrafish model of neutrophilic inflammation and promoted erythroid differentiation and GATA1 accumulation in K562 cells. In conclusion, our results reveal that the NLRP1 inflammasome regulates hematopoiesis and pave the way to develop novel therapeutic strategies for the treatment of hematopoietic alterations associated with chronic inflammatory and rare diseases.
Collapse
Affiliation(s)
- Lola Rodríguez‐Ruiz
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - Juan M Lozano‐Gil
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - Elena Naranjo‐Sánchez
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
- Hospital Clínico Universitario Virgen de la ArrixacaMurciaSpain
| | - Elena Martínez‐Balsalobre
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
- Hospital Clínico Universitario Virgen de la ArrixacaMurciaSpain
| | - Alicia Martínez‐López
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - Christophe Lachaud
- Aix‐Marseille University, Inserm, CNRS, Institut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Miguel Blanquer
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Hospital Clínico Universitario Virgen de la ArrixacaMurciaSpain
- Departamento de Medicina y Unidad de Terapia Celular y Trasplante Hematopoyético, Facultad de MedicinaUniversidad de MurciaMurciaSpain
| | - Toan K Phung
- MRC PPU, Sir James Black Centre, School of Life SciencesUniversity of DundeeDundeeUK
| | - Diana García‐Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - María L Cayuela
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
- Hospital Clínico Universitario Virgen de la ArrixacaMurciaSpain
| | - Sylwia D Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - Ana B Pérez‐Oliva
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de BiologíaUniversidad de MurciaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria (IMIB)‐Pascual ParrillaMurciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
14
|
Martínez-López A, Candel S, Tyrkalska SD. Animal models of silicosis: fishing for new therapeutic targets and treatments. Eur Respir Rev 2023; 32:230078. [PMID: 37558264 PMCID: PMC10424253 DOI: 10.1183/16000617.0078-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023] Open
Abstract
Silicosis as an occupational lung disease has been present in our lives for centuries. Research studies have already developed and implemented many animal models to study the pathogenesis and molecular basis of the disease and enabled the search for treatments. As all experimental animal models used to date have their advantages and disadvantages, there is a continuous search for a better model, which will not only accelerate basic research, but also contribute to clinical aspects and drug development. We review here, for the first time, the main animal models developed to date to study silicosis and the unique advantages of the zebrafish model that make it an optimal complement to other models. Among the main advantages of zebrafish for modelling human diseases are its ease of husbandry, low maintenance cost, external fertilisation and development, its transparency from early life, and its amenability to chemical and genetic screening. We discuss the use of zebrafish as a model of silicosis, its similarities to other animal models and the characteristics of patients at molecular and clinical levels, and show the current state of the art of inflammatory and fibrotic zebrafish models that could be used in silicosis research.
Collapse
Affiliation(s)
- Alicia Martínez-López
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- These authors contributed equally to this work
| | - Sergio Candel
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
- These authors contributed equally to this work
| | - Sylwia D Tyrkalska
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
15
|
Faught E, Schaaf MJM. The Mineralocorticoid Receptor Plays a Crucial Role in Macrophage Development and Function. Endocrinology 2023; 164:bqad127. [PMID: 37597174 PMCID: PMC10475750 DOI: 10.1210/endocr/bqad127] [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: 05/16/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023]
Abstract
Stress and the attendant rise in glucocorticoids (GCs) results in a potent suppression of the immune system. To date, the anti-inflammatory role of GCs, via activation of the glucocorticoid receptor, has been well-characterized. However, cortisol, the primary GC in both fish and humans, also signals through the high-affinity mineralocorticoid receptor (MR), of which the immunomodulatory role is poorly understood. Here, we tested the hypothesis that MR is a key modulator of leukocyte function during inflammation. Using transgenic MR knockout zebrafish with fluorescently labelled leukocytes, we show that a loss of MR results in a global reduction in macrophage number during key development stages. This reduction was associated with impaired macrophage proliferation and responsivity to developmental distribution signals, as well as increased susceptibility to cell death. Using a tail fin amputation in zebrafish larvae as a model for localized inflammation, we further showed that MR knockout larvae display a reduced ability to produce more macrophages under periods of inflammation (emergency myelopoiesis). Finally, we treated wild-type larvae with an MR antagonist (eplerenone) during definitive hematopoiesis, when the macrophages had differentiated normally throughout the larvae. This pharmacological blockade of MR reduced the migration of macrophages toward a wound, which was associated with reduced macrophage Ccr2 signalling. Eplerenone treatment also abolished the cortisol-induced inhibition of macrophage migration, suggesting a role for MR in cortisol-mediated anti-inflammatory action. Taken together, our work reveals that MR is a key modulator of the innate immune response to inflammation under both basal and stressed conditions.
Collapse
Affiliation(s)
- Erin Faught
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| | - Marcel J M Schaaf
- Institute of Biology Leiden, Leiden University, Leiden 2333CC, The Netherlands
| |
Collapse
|
16
|
Lucianò AM, Di Martile M, Pérez-Oliva AB, Di Caprio M, Foddai ML, Buglioni S, Mulero V, Del Bufalo D. Exploring association of melanoma-specific Bcl-xL with tumor immune microenvironment. J Exp Clin Cancer Res 2023; 42:178. [PMID: 37488586 PMCID: PMC10364435 DOI: 10.1186/s13046-023-02735-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/16/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Macrophages take center stage in the tumor microenvironment, a niche composed of extracellular matrix and a heterogeneous group of cells, including immune ones. They can evolve during tumor progression and acquire Tumor-Associated Macrophage (TAMs) phenotype. The release of cytokines by tumor and stromal cells, influence the secretion of cytokines by TAMs, which can guarantee tumor progression and influence the response to therapy. Among all factors able to recruit and polarize macrophages, we focused our attention on Bcl-xL, a multifaceted member of the Bcl-2 family, whose expression is deregulated in melanoma. It acts not only as a canonical pro-survival and anti-apoptotic protein, but also as a promoter of tumor progression. METHODS Human melanoma cells silencing or overexpressing Bcl-xL protein, THP-1 monocytic cells and monocyte-derived macrophages were used in this study. Protein array and specific neutralizing antibodies were used to analyze cytokines and chemokines secreted by melanoma cells. qRT-PCR, ELISA and Western Blot analyses were used to evaluate macrophage polarization markers and protein expression levels. Transwell chambers were used to evaluate migration of THP-1 and monocyte-derived macrophages. Mouse and zebrafish models were used to evaluate the ability of melanoma cells to recruit and polarize macrophages in vivo. RESULTS We demonstrated that melanoma cells overexpressing Bcl-xL recruit macrophages at the tumor site and induce a M2 phenotype. In addition, we identified that interleukin-8 and interleukin-1β cytokines are involved in macrophage polarization, and the chemokine CCL5/RANTES in the macrophages recruitment at the tumor site. We also found that all these Bcl-xL-induced factors are regulated in a NF-kB dependent manner in human and zebrafish melanoma models. CONCLUSIONS Our findings confirmed the pro-tumoral function of Bcl-xL in melanoma through its effects on macrophage phenotype.
Collapse
Affiliation(s)
- Anna Maria Lucianò
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, 30100, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Ana B Pérez-Oliva
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain
| | - Marica Di Caprio
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Laura Foddai
- Immunohematology and Transfusional Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Simonetta Buglioni
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, 30100, Spain.
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, 28029, Spain.
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
17
|
Dinarello A, Betto RM, Diamante L, Tesoriere A, Ghirardo R, Cioccarelli C, Meneghetti G, Peron M, Laquatra C, Tiso N, Martello G, Argenton F. STAT3 and HIF1α cooperatively mediate the transcriptional and physiological responses to hypoxia. Cell Death Discov 2023; 9:226. [PMID: 37407568 DOI: 10.1038/s41420-023-01507-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/04/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
STAT3 and HIF1α are two fundamental transcription factors involved in many merging processes, like angiogenesis, metabolism, and cell differentiation. Notably, under pathological conditions, the two factors have been shown to interact genetically, but both the molecular mechanisms underlying such interactions and their relevance under physiological conditions remain unclear. In mouse embryonic stem cells (ESCs) we manage to determine the specific subset of hypoxia-induced genes that need STAT3 to be properly transcribed and, among them, fundamental genes like Vegfa, Hk1, Hk2, Pfkp and Hilpda are worth mentioning. Unexpectedly, we also demonstrated that the absence of STAT3 does not affect the expression of Hif1α mRNA nor the stabilization of HIF1α protein, but the STAT3-driven regulation of the hypoxia-dependent subset of gene could rely on the physical interaction between STAT3 and HIF1α. To further elucidate the physiological roles of this STAT3 non-canonical nuclear activity, we used a CRISPR/Cas9 zebrafish stat3 knock-out line. Notably, hypoxia-related fluorescence of the hypoxia zebrafish reporter line (HRE:mCherry) cannot be induced when Stat3 is not active and, while Stat3 Y705 phosphorylation seems to have a pivotal role in this process, S727 does not affect the Stat3-dependent hypoxia response. Hypoxia is fundamental for vascularization, angiogenesis and immune cells mobilization; all processes that, surprisingly, cannot be induced by low oxygen levels when Stat3 is genetically ablated. All in all, here we report the specific STAT3/HIF1α-dependent subset of genes in vitro and, for the first time with an in vivo model, we determined some of the physiological roles of STAT3-hypoxia crosstalk.
Collapse
Affiliation(s)
| | | | - Linda Diamante
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | | | | | | | - Claudio Laquatra
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | | | | |
Collapse
|
18
|
Yu T, Kuang H, Wu X, Huang Y, Wang J, Wen Z. Cell competition for neuron-derived trophic factor controls the turnover and lifespan of microglia. SCIENCE ADVANCES 2023; 9:eadf9790. [PMID: 37327343 PMCID: PMC10275588 DOI: 10.1126/sciadv.adf9790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/10/2023] [Indexed: 06/18/2023]
Abstract
Microglia are brain-resident macrophages capable of long-term maintenance through self-renewal. Yet the mechanism governing the turnover and lifespan of microglia remains unknown. In zebrafish, microglia arise from two sources, rostral blood island (RBI) and aorta-gonad-mesonephros (AGM). The RBI-derived microglia are born early but have a short lifespan and diminish in adulthood, while the AGM-derived microglia emerge later and are capable of long-term maintenance in adulthood. Here, we show that the attenuation of RBI microglia is due to their less competitiveness for neuron-derived interleukin-34 (Il34) caused by age-dependent decline of colony-stimulating factor-1 receptor a (csf1ra). Alterations of Il34/Csf1ra levels and removal of AGM microglia revamp the proportion and lifespan of RBI microglia. The csf1ra/CSF1R expression in zebrafish AGM-derived microglia and murine adult microglia also undergo age-dependent decline, leading to the elimination of aged microglia. Our study reveals cell competition as a general mechanism controlling the turnover and lifespan of microglia.
Collapse
Affiliation(s)
- Tao Yu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Haoyue Kuang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Xiaohai Wu
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Ying Huang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Jianzhong Wang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Zilong Wen
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
19
|
Emmerich K, White DT, Kambhampati SP, Casado GL, Fu TM, Chunawala Z, Sahoo A, Nimmagadda S, Krishnan N, Saxena MT, Walker SL, Betzig E, Kannan RM, Mumm JS. Nanoparticle-based targeting of microglia improves the neural regeneration enhancing effects of immunosuppression in the zebrafish retina. Commun Biol 2023; 6:534. [PMID: 37202450 PMCID: PMC10193316 DOI: 10.1038/s42003-023-04898-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Abstract
Retinal Müller glia function as injury-induced stem-like cells in zebrafish but not mammals. However, insights gleaned from zebrafish have been applied to stimulate nascent regenerative responses in the mammalian retina. For instance, microglia/macrophages regulate Müller glia stem cell activity in the chick, zebrafish, and mouse. We previously showed that post-injury immunosuppression by the glucocorticoid dexamethasone accelerated retinal regeneration kinetics in zebrafish. Similarly, microglia ablation enhances regenerative outcomes in the mouse retina. Targeted immunomodulation of microglia reactivity may therefore enhance the regenerative potential of Müller glia for therapeutic purposes. Here, we investigated potential mechanisms by which post-injury dexamethasone accelerates retinal regeneration kinetics, and the effects of dendrimer-based targeting of dexamethasone to reactive microglia. Intravital time-lapse imaging revealed that post-injury dexamethasone inhibited microglia reactivity. The dendrimer-conjugated formulation: (1) decreased dexamethasone-associated systemic toxicity, (2) targeted dexamethasone to reactive microglia, and (3) improved the regeneration enhancing effects of immunosuppression by increasing stem/progenitor proliferation rates. Lastly, we show that the gene rnf2 is required for the enhanced regeneration effect of D-Dex. These data support the use of dendrimer-based targeting of reactive immune cells to reduce toxicity and enhance the regeneration promoting effects of immunosuppressants in the retina.
Collapse
Affiliation(s)
- Kevin Emmerich
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - David T White
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Siva P Kambhampati
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Grace L Casado
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Tian-Ming Fu
- Janelia Farms Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Electrical and Computer Engineering and Princeton Bioengineering Initiative, Princeton University, Princeton, NJ, USA
| | - Zeeshaan Chunawala
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Arpan Sahoo
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Saumya Nimmagadda
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Nimisha Krishnan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Steven L Walker
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Eric Betzig
- Janelia Farms Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Rangaramanujam M Kannan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Jeff S Mumm
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
20
|
Peterman E, Quitevis EJA, Black EC, Horton EC, Aelmore RL, White E, Sagasti A, Rasmussen JP. Zebrafish cutaneous injury models reveal that Langerhans cells engulf axonal debris in adult epidermis. Dis Model Mech 2023; 16:dmm049911. [PMID: 36876992 PMCID: PMC10110399 DOI: 10.1242/dmm.049911] [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: 09/29/2022] [Accepted: 02/28/2023] [Indexed: 03/07/2023] Open
Abstract
Somatosensory neurons extend enormous peripheral axons to the skin, where they detect diverse environmental stimuli. Somatosensory peripheral axons are easily damaged due to their small caliber and superficial location. Axonal damage results in Wallerian degeneration, creating vast quantities of cellular debris that phagocytes must remove to maintain organ homeostasis. The cellular mechanisms that ensure efficient clearance of axon debris from stratified adult skin are unknown. Here, we established zebrafish scales as a tractable model to study axon degeneration in the adult epidermis. Using this system, we demonstrated that skin-resident immune cells known as Langerhans cells engulf the majority of axon debris. In contrast to immature skin, adult keratinocytes did not significantly contribute to debris removal, even in animals lacking Langerhans cells. Our study establishes a powerful new model for studying Wallerian degeneration and identifies a new function for Langerhans cells in maintenance of adult skin homeostasis following injury. These findings have important implications for pathologies that trigger somatosensory axon degeneration.
Collapse
Affiliation(s)
- Eric Peterman
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | | | - Erik C. Black
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Emma C. Horton
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Rune L. Aelmore
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Ethan White
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Alvaro Sagasti
- Molecular, Cell and Developmental Biology Department, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Jeffrey P. Rasmussen
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
21
|
Leiba J, Özbilgiç R, Hernández L, Demou M, Lutfalla G, Yatime L, Nguyen-Chi M. Molecular Actors of Inflammation and Their Signaling Pathways: Mechanistic Insights from Zebrafish. BIOLOGY 2023; 12:153. [PMID: 36829432 PMCID: PMC9952950 DOI: 10.3390/biology12020153] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Inflammation is a hallmark of the physiological response to aggressions. It is orchestrated by a plethora of molecules that detect the danger, signal intracellularly, and activate immune mechanisms to fight the threat. Understanding these processes at a level that allows to modulate their fate in a pathological context strongly relies on in vivo studies, as these can capture the complexity of the whole process and integrate the intricate interplay between the cellular and molecular actors of inflammation. Over the years, zebrafish has proven to be a well-recognized model to study immune responses linked to human physiopathology. We here provide a systematic review of the molecular effectors of inflammation known in this vertebrate and recapitulate their modes of action, as inferred from sterile or infection-based inflammatory models. We present a comprehensive analysis of their sequence, expression, and tissue distribution and summarize the tools that have been developed to study their function. We further highlight how these tools helped gain insights into the mechanisms of immune cell activation, induction, or resolution of inflammation, by uncovering downstream receptors and signaling pathways. These progresses pave the way for more refined models of inflammation, mimicking human diseases and enabling drug development using zebrafish models.
Collapse
|
22
|
Characterization of the innate immune response to Streptococcus pneumoniae infection in zebrafish. PLoS Genet 2023; 19:e1010586. [PMID: 36622851 PMCID: PMC9858863 DOI: 10.1371/journal.pgen.1010586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/20/2023] [Accepted: 12/20/2022] [Indexed: 01/10/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is one of the most frequent causes of pneumonia, sepsis and meningitis in humans, and an important cause of mortality among children and the elderly. We have previously reported the suitability of the zebrafish (Danio rerio) larval model for the study of the host-pathogen interactions in pneumococcal infection. In the present study, we characterized the zebrafish innate immune response to pneumococcus in detail through a whole-genome level transcriptome analysis and revealed a well-conserved response to this human pathogen in challenged larvae. In addition, to gain understanding of the genetic factors associated with the increased risk for severe pneumococcal infection in humans, we carried out a medium-scale forward genetic screen in zebrafish. In the screen, we identified a mutant fish line which showed compromised resistance to pneumococcus in the septic larval infection model. The transcriptome analysis of the mutant zebrafish larvae revealed deficient expression of a gene homologous for human C-reactive protein (CRP). Furthermore, knockout of one of the six zebrafish crp genes by CRISPR-Cas9 mutagenesis predisposed zebrafish larvae to a more severe pneumococcal infection, and the phenotype was further augmented by concomitant knockdown of a gene for another Crp isoform. This suggests a conserved function of C-reactive protein in anti-pneumococcal immunity in zebrafish. Altogether, this study highlights the similarity of the host response to pneumococcus in zebrafish and humans, gives evidence of the conserved role of C-reactive protein in the defense against pneumococcus, and suggests novel host genes associated with pneumococcal infection.
Collapse
|
23
|
Tyrkalska SD, Pedoto A, Martínez-López A, Ros-Lucas JA, Mesa-Del-Castillo P, Candel S, Mulero V. Silica crystals activate toll-like receptors and inflammasomes to promote local and systemic immune responses in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104523. [PMID: 36055417 DOI: 10.1016/j.dci.2022.104523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Silica crystals are potent activators of the inflammasome that cause a fibrotic lung disease, called silicosis, with no effective treatment available. We report here that injection of silica crystals into the hindbrain ventricle of zebrafish embryos led to the initiation of local and systemic immune responses driven through both Toll-like receptors (TLR)- and inflammasome-dependent signaling pathways, followed by induction of pro-fibrotic markers. Genetic and pharmacological analysis revealed that the Nlrp3 inflammasome regulated silica crystal-induced inflammation and pyroptotic cell death, but not emergency myelopoiesis. In addition, Cxcl8a/Cxcr2-dependent recruitment of myeloid cells to silica crystals was required to promote emergency myelopoiesis and systemic inflammation. The zebrafish model of silicosis developed here shed light onto the molecular mechanisms involved in the activation of the immune system by silica crystals.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Annamaria Pedoto
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Alicia Martínez-López
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - José A Ros-Lucas
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Servicio de Neumología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Pablo Mesa-Del-Castillo
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain; Servicio de Reumatología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Sergio Candel
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
24
|
Macrophage NFATC2 mediates angiogenic signaling during mycobacterial infection. Cell Rep 2022; 41:111817. [PMID: 36516756 PMCID: PMC9880963 DOI: 10.1016/j.celrep.2022.111817] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
During mycobacterial infections, pathogenic mycobacteria manipulate both host immune and stromal cells to establish and maintain a productive infection. In humans, non-human primates, and zebrafish models of infection, pathogenic mycobacteria produce and modify the specialized lipid trehalose 6,6'-dimycolate (TDM) in the bacterial cell envelope to drive host angiogenesis toward the site of forming granulomas, leading to enhanced bacterial growth. Here, we use the zebrafish-Mycobacterium marinum infection model to define the signaling basis of the host angiogenic response. Through intravital imaging and cell-restricted peptide-mediated inhibition, we identify macrophage-specific activation of NFAT signaling as essential to TDM-mediated angiogenesis in vivo. Exposure of cultured human cells to Mycobacterium tuberculosis results in robust induction of VEGFA, which is dependent on a signaling pathway downstream of host TDM detection and culminates in NFATC2 activation. As granuloma-associated angiogenesis is known to serve bacterial-beneficial roles, these findings identify potential host targets to improve tuberculosis disease outcomes.
Collapse
|
25
|
Saelens JW, Sweeney MI, Viswanathan G, Xet-Mull AM, Jurcic Smith KL, Sisk DM, Hu DD, Cronin RM, Hughes EJ, Brewer WJ, Coers J, Champion MM, Champion PA, Lowe CB, Smith CM, Lee S, Stout JE, Tobin DM. An ancestral mycobacterial effector promotes dissemination of infection. Cell 2022; 185:4507-4525.e18. [PMID: 36356582 PMCID: PMC9691622 DOI: 10.1016/j.cell.2022.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/27/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.
Collapse
Affiliation(s)
- Joseph W Saelens
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mollie I Sweeney
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ana María Xet-Mull
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kristen L Jurcic Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dana M Sisk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel D Hu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel M Cronin
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sunhee Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jason E Stout
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Division of Infectious Diseases and International Health, Duke University School of Medicine, Durham, NC 27710, USA.
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
26
|
Uricase-Deficient Larval Zebrafish with Elevated Urate Levels Demonstrate Suppressed Acute Inflammatory Response to Monosodium Urate Crystals and Prolonged Crystal Persistence. Genes (Basel) 2022; 13:genes13122179. [PMID: 36553446 PMCID: PMC9777727 DOI: 10.3390/genes13122179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Gout is caused by elevated serum urate leading to the deposition of monosodium urate (MSU) crystals that can trigger episodes of acute inflammation. Humans are sensitive to developing gout because they lack a functional urate-metabolizing enzyme called uricase/urate oxidase (encoded by the UOX gene). A hallmark of long-standing disease is tophaceous gout, characterized by the formation of tissue-damaging granuloma-like structures ('tophi') composed of densely packed MSU crystals and immune cells. Little is known about how tophi form, largely due to the lack of suitable animal models in which the host response to MSU crystals can be studied in vivo long-term. We have previously described a larval zebrafish model of acute gouty inflammation where the host response to microinjected MSU crystals can be live imaged within an intact animal. Although useful for modeling acute inflammation, crystals are rapidly cleared following a robust innate immune response, precluding analysis at later stages. Here we describe a zebrafish uox null mutant that possesses elevated urate levels at larval stages. Uricase-deficient 'hyperuricemic' larvae exhibit a suppressed acute inflammatory response to MSU crystals and prolonged in vivo crystal persistence. Imaging of crystals at later stages reveals that they form granuloma-like structures dominated by macrophages. We believe that uox-/- larvae will provide a useful tool to explore the transition from acute gouty inflammation to tophus formation, one of the remaining mysteries of gout pathogenesis.
Collapse
|
27
|
Pagán AJ, Lee LJ, Edwards-Hicks J, Moens CB, Tobin DM, Busch-Nentwich EM, Pearce EL, Ramakrishnan L. mTOR-regulated mitochondrial metabolism limits mycobacterium-induced cytotoxicity. Cell 2022; 185:3720-3738.e13. [PMID: 36103894 PMCID: PMC9596383 DOI: 10.1016/j.cell.2022.08.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/17/2022] [Accepted: 08/16/2022] [Indexed: 02/01/2023]
Abstract
Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.
Collapse
Affiliation(s)
- Antonio J. Pagán
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK,Department of Microbiology, University of Washington, Seattle, WA 98195, USA,Corresponding author
| | - Lauren J. Lee
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Cecilia B. Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - David M. Tobin
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Elisabeth M. Busch-Nentwich
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Erika L. Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK,Department of Microbiology, University of Washington, Seattle, WA 98195, USA,Corresponding author
| |
Collapse
|
28
|
Mafba and Mafbb regulate microglial colonization of zebrafish brain via controlling chemotaxis receptor expression. Proc Natl Acad Sci U S A 2022; 119:e2203273119. [PMID: 36122226 PMCID: PMC9522419 DOI: 10.1073/pnas.2203273119] [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] [Indexed: 11/25/2022] Open
Abstract
Microglia are a subpopulation of macrophages residing in the central nervous system (CNS). Because microglial precursors/peripheral macrophages are born in peripheral hematopoietic tissues, the establishment of a microglia pool in the CNS involves two processes: colonization, the homing of macrophages from peripheral tissues to the CNS, and maturation, the differentiation of brain-colonizing macrophages into microglia. This study aims to investigate the molecular mechanisms underlying microglial colonization during early development. Utilizing a zebrafish model system, we show that Mafba and Mafbb, two zebrafish orthologs of mammalian MAFB essential for macrophage differentiation and phagocytosis, regulate microglial colonization of the brain via modulating the lysoPS-Gpr34a signaling pathway during early embryogenesis. Our findings reveal a previously unappreciated genetic mechanism involved in microglial colonization of the brain. Microglia are the central nervous system (CNS)–resident macrophages involved in neural inflammation, neurogenesis, and neural activity regulation. Previous studies have shown that naturally occurring neuronal apoptosis plays a critical role in regulating microglial colonization of the brain in zebrafish. However, the molecular signaling cascades underlying neuronal apoptosis-mediated microglial colonization and the regulation of these cascades remain undefined. Here, we show that basic leucine zipper (b-Zip) transcription factors, Mafba and Mafbb, two zebrafish orthologs of mammalian MAFB, are key regulators in neuronal apoptosis-mediated microglial colonization of the brain in zebrafish. We document that the loss of Mafba and Mafbb function perturbs microglial colonization of the brain. We further demonstrate that Mafba and Mafbb act cell-autonomously and cooperatively to orchestrate microglial colonization, at least in part, by regulating the expression of G protein–coupled receptor 34a (Gpr34a), which directs peripheral macrophage recruitment into the brain through sensing the lysophosphatidylserine (lysoPS) released by the apoptotic neurons. Our study reveals that Mafba and Mafbb regulate neuronal apoptosis-mediated microglial colonization of the brain in zebrafish via the lysoPS-Gpr34a pathway.
Collapse
|
29
|
Lou Demy D, Touret AL, Lancino M, Tauzin M, Capuana L, Pierre C, Herbomel P. Trim33 conditions the lifespan of primitive macrophages and onset of definitive macrophage production. Development 2022; 149:276505. [DOI: 10.1242/dev.200835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Trim33 (Tif1γ) is a transcriptional regulator that is notably involved in several aspects of hematopoiesis. It is essential for the production of erythrocytes in zebrafish, and for the proper functioning and aging of hematopoietic stem and progenitor cells (HSPCs) in mice. Here, we have found that, in zebrafish development, Trim33 is essential cell-autonomously for the lifespan of the yolk sac-derived primitive macrophages, as well as for the initial production of definitive (HSPC-derived) macrophages in the first niche of definitive hematopoiesis, the caudal hematopoietic tissue. Moreover, Trim33 deficiency leads to an excess production of definitive neutrophils and thrombocytes. Our data indicate that Trim33 radically conditions the differentiation output of aorta-derived HSPCs in all four erythro-myeloid cell types, in a niche-specific manner.
Collapse
Affiliation(s)
- Doris Lou Demy
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Anne-Lou Touret
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Mylène Lancino
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Muriel Tauzin
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Lavinia Capuana
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Constance Pierre
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Philippe Herbomel
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| |
Collapse
|
30
|
Rubin SA, Baron CS, Pessoa Rodrigues C, Duran M, Corbin AF, Yang SP, Trapnell C, Zon LI. Single-cell analyses reveal early thymic progenitors and pre-B cells in zebrafish. J Exp Med 2022; 219:e20220038. [PMID: 35938989 PMCID: PMC9365674 DOI: 10.1084/jem.20220038] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/11/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
The zebrafish has proven to be a valuable model organism for studying hematopoiesis, but relatively little is known about zebrafish immune cell development and functional diversity. Elucidating key aspects of zebrafish lymphocyte development and exploring the breadth of effector functions would provide valuable insight into the evolution of adaptive immunity. We performed single-cell RNA sequencing on ∼70,000 cells from the zebrafish marrow and thymus to establish a gene expression map of zebrafish immune cell development. We uncovered rich cellular diversity in the juvenile and adult zebrafish thymus, elucidated B- and T-cell developmental trajectories, and transcriptionally characterized subsets of hematopoietic stem and progenitor cells and early thymic progenitors. Our analysis permitted the identification of two dendritic-like cell populations and provided evidence in support of the existence of a pre-B cell state. Our results provide critical insights into the landscape of zebrafish immunology and offer a foundation for cellular and genetic studies.
Collapse
Affiliation(s)
- Sara A. Rubin
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA
| | - Chloé S. Baron
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA
| | - Cecilia Pessoa Rodrigues
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA
| | - Madeleine Duran
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Alexandra F. Corbin
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
| | - Song P. Yang
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Boston, MA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
31
|
Iribarne M, Hyde DR. Different inflammation responses modulate Müller glia proliferation in the acute or chronically damaged zebrafish retina. Front Cell Dev Biol 2022; 10:892271. [PMID: 36120571 PMCID: PMC9472244 DOI: 10.3389/fcell.2022.892271] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Unlike mammals, zebrafish regenerate in response to retinal damage. Because microglia are activated by retinal damage, we investigated their role during regeneration following either acute or chronic damage. At three weeks post-fertilization (wpf), both wild-type fish exhibiting NMDA-induced acute ganglion and amacrine cell death and gold rush (gosh) mutant fish possessing chronic cone photoreceptor degeneration displayed reactive microglia/macrophages and Müller glia proliferation. Dexamethasone-treated retinas, to inhibit the immune response, lacked reactive microglia/macrophages and possessed fewer PCNA-positive cells, while LPS treatment increased microglia/macrophages and PCNA-labeled cells. NMDA-injured retinas upregulated expression of il-1β and tnfα pro-inflammatory cytokine genes, followed by increased expression of il-10 and arg1 anti-inflammatory/remodeling cytokine genes. A transient early TNFα pro-inflammatory microglia/macrophage population was visualized in NMDA-damaged retinas. In contrast, gosh mutant retinas exhibited a slight increase of pro-inflammatory cytokine gene expression concurrently with a greater increased anti-inflammatory/remodeling cytokine gene expression. Few TNFα pro-inflammatory microglia/macrophages were observed in the gosh retina. Understanding why acute and chronic damage results in different inflammation profiles and their effects on regulating zebrafish retinal regeneration would provide important clues toward improving therapeutic strategies for repairing injured mammalian tissues.
Collapse
Affiliation(s)
- Maria Iribarne
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
| | - David R. Hyde
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
- *Correspondence: David R. Hyde,
| |
Collapse
|
32
|
Mohammadi A, Sorensen GL, Pilecki B. MFAP4-Mediated Effects in Elastic Fiber Homeostasis, Integrin Signaling and Cancer, and Its Role in Teleost Fish. Cells 2022; 11:cells11132115. [PMID: 35805199 PMCID: PMC9265350 DOI: 10.3390/cells11132115] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Microfibrillar-associated protein 4 (MFAP4) is an extracellular matrix (ECM) protein belonging to the fibrinogen-related domain superfamily. MFAP4 is highly expressed in elastin-rich tissues such as lung, blood vessels and skin. MFAP4 is involved in organization of the ECM, regulating proper elastic fiber assembly. On the other hand, during pathology MFAP4 actively contributes to disease development and progression due to its interactions with RGD-dependent integrin receptors. Both tissue expression and circulating MFAP4 levels are associated with various disorders, including liver fibrosis and cancer. In other experimental models, such as teleost fish, MFAP4 appears to participate in host defense as a macrophage-specific innate immune molecule. The aim of this review is to summarize the accumulating evidence that indicates the importance of MFAP4 in homeostasis as well as pathological conditions, discuss its known biological functions with special focus on elastic fiber assembly, integrin signaling and cancer, as well as describe the reported functions of non-mammalian MFAP4 in fish. Overall, our work provides a comprehensive overview on the role of MFAP4 in health and disease.
Collapse
|
33
|
Pardo-Sánchez I, García-Moreno D, Mulero V. Zebrafish Models to Study the Crosstalk between Inflammation and NADPH Oxidase-Derived Oxidative Stress in Melanoma. Antioxidants (Basel) 2022; 11:1277. [PMID: 35883768 PMCID: PMC9311651 DOI: 10.3390/antiox11071277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer, and its incidence continues to increase. In the early stages of melanoma, when the malignant cells have not spread to lymph nodes, they can be removed by simple surgery and there is usually low recurrence. Melanoma has a high mortality rate due to its ability to metastasize; once melanoma has spread, it becomes a major health complication. For these reasons, it is important to study how healthy melanocytes transform into melanoma cells, how they interact with the immune system, which mechanisms they use to escape immunosurveillance, and, finally, how they spread and colonize other tissues, metastasizing. Inflammation and oxidative stress play important roles in the development of several types of cancer, including melanoma, but it is not yet clear under which conditions they are beneficial or detrimental. Models capable of studying the relevance of inflammation and oxidative stress in the early steps of melanocyte transformation are urgently needed, as they are expected to help recognize premetastatic lesions in patients by improving both early detection and the development of new therapies.
Collapse
Affiliation(s)
- Irene Pardo-Sánchez
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Diana García-Moreno
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
34
|
Chen L, Su B, Yu J, Wang J, Hu H, Ren HQ, Wu B. Combined effects of arsenic and 2,2-dichloroacetamide on different cell populations of zebrafish liver. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:152961. [PMID: 35031379 DOI: 10.1016/j.scitotenv.2022.152961] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 06/14/2023]
Abstract
Arsenic (As) and disinfection by-products are important health risk factors in the water environment. However, their combined effects on different cell populations in the liver are not well known. Here, zebrafish were exposed to 100 μg/L As, 300 μg/L 2,2-dichloroacetamide (DCAcAm), and their combination for 23 days. Then transcriptome profiles of cell populations in zebrafish liver were analyzed by single-cell RNA sequencing (scRNA-seq). A total of 13,563 cells were obtained, which were identified as hepatocytes, hepatic duct cells, endothelial cells and macrophages. Hepatocytes were the main target cell subtype of As and DCAcAm exposures. DCAcAm exposure induced higher toxicity in male hepatocytes, which specifically changed amino acid metabolism, response to hormone and cofactor metabolism. However, As exposure caused higher toxicity in female hepatocytes, which altered lipid metabolism, carbon metabolism, and peroxisome. Combined exposure to As and DCAcAm decreased toxicities in hepatocytes compared to each one alone. Female hepatocytes had higher tolerance to co-exposure of As and DCAcAm than male hepatocytes. Further, combined exposure to As and DCAcAm induced functional changes in macrophages similar to As alone groups, which mainly altered the transfer of sterol and cholesterol. Hepatic duct cells and endothelial cells were not influenced by exposures to As and DCAcAm. This study for the first time highlights the cell-specific combined responses of As and DCAcAm in zebrafish liver, which provide useful information for their health risk assessment in a co-exposure environment.
Collapse
Affiliation(s)
- Ling Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Bei Su
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Jing Yu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Jinfeng Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Haidong Hu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Hong-Qiang Ren
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
35
|
Kanaan R, Medlej-Hashim M, Jounblat R, Pilecki B, Sorensen GL. Microfibrillar-associated protein 4 in health and disease. Matrix Biol 2022; 111:1-25. [DOI: 10.1016/j.matbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
36
|
Geurtzen K, López-Delgado AC, Duseja A, Kurzyukova A, Knopf F. Laser-mediated osteoblast ablation triggers a pro-osteogenic inflammatory response regulated by reactive oxygen species and glucocorticoid signaling in zebrafish. Development 2022; 149:275194. [DOI: 10.1242/dev.199803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022]
Abstract
ABSTRACT
In zebrafish, transgenic labeling approaches, robust regenerative responses and excellent in vivo imaging conditions enable precise characterization of immune cell behavior in response to injury. Here, we monitored osteoblast-immune cell interactions in bone, a tissue which is particularly difficult to in vivo image in tetrapod species. Ablation of individual osteoblasts leads to recruitment of neutrophils and macrophages in varying numbers, depending on the extent of the initial insult, and initiates generation of cathepsin K+ osteoclasts from macrophages. Osteoblast ablation triggers the production of pro-inflammatory cytokines and reactive oxygen species, which are needed for successful macrophage recruitment. Excess glucocorticoid signaling as it occurs during the stress response inhibits macrophage recruitment, maximum speed and changes the macrophage phenotype. Although osteoblast loss is compensated for within a day by contribution of committed osteoblasts, macrophages continue to populate the region. Their presence is required for osteoblasts to fill the lesion site. Our model enables visualization of bone repair after microlesions at single-cell resolution and demonstrates a pro-osteogenic function of tissue-resident macrophages in non-mammalian vertebrates.
Collapse
Affiliation(s)
- Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ankita Duseja
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Department of Oncology and Metabolism, Metabolic Bone Centre, Sorby Wing, Northern General Hospital, Sheffield S5 7AU, UK
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Faculty of Health and Medical Sciences, Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
37
|
Bohaud C, Johansen MD, Varga B, Contreras-Lopez R, Barthelaix A, Hamela C, Sapède D, Cloitre T, Gergely C, Jorgensen C, Kremer L, Djouad F. Exploring Macrophage-Dependent Wound Regeneration During Mycobacterial Infection in Zebrafish. Front Immunol 2022; 13:838425. [PMID: 35401552 PMCID: PMC8987025 DOI: 10.3389/fimmu.2022.838425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
The molecular and cellular mechanisms associated with tissue degradation or regeneration in an infectious context are poorly defined. Herein, we explored the role of macrophages in orchestrating either tissue regeneration or degradation in zebrafish embryos pre-infected with the fish pathogen Mycobacterium marinum. Zebrafish were inoculated with different infectious doses of M. marinum prior to fin resection. While mild infection accelerated fin regeneration, moderate or severe infection delayed this process by reducing blastemal cell proliferation and impeding tissue morphogenesis. This was correlated with impaired macrophage recruitment at the wound of the larvae receiving high infectious doses. Macrophage activation characterized, in part, by a high expression level of tnfa was exacerbated in severely infected fish during the early phase of the regeneration process, leading to macrophage necrosis and their complete absence in the later phase. Our results demonstrate how a mycobacterial infection influences the macrophage response and tissue regenerative processes.
Collapse
Affiliation(s)
| | - Matt D. Johansen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Béla Varga
- L2C, Univ Montpellier, CNRS, Montpellier, France
| | | | | | - Claire Hamela
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Dora Sapède
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | | | | | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | - Farida Djouad
- IRMB, Univ Montpellier, INSERM, Montpellier, France
- *Correspondence: Farida Djouad,
| |
Collapse
|
38
|
Robertson TF, Huttenlocher A. Real-time imaging of inflammation and its resolution: It's apparent because it's transparent. Immunol Rev 2022; 306:258-270. [PMID: 35023170 PMCID: PMC8855992 DOI: 10.1111/imr.13061] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
The ability to directly observe leukocyte behavior in vivo has dramatically expanded our understanding of the immune system. Zebrafish are particularly amenable to the high-resolution imaging of leukocytes during both homeostasis and inflammation. Due to its natural transparency, intravital imaging in zebrafish does not require any surgical manipulation. As a result, zebrafish are particularly well-suited for the long-term imaging required to observe the temporal and spatial events during the onset and resolution of inflammation. Here, we review major insights about neutrophil and macrophage function gained from real-time imaging of zebrafish. We discuss neutrophil reverse migration, the process whereby neutrophils leave sites of tissue damage and resolve local inflammation. Further, we discuss the current tools available for investigating immune function in zebrafish and how future studies that simultaneously image multiple leukocyte subsets can be used to further dissect mechanisms that regulate both the onset and resolution of inflammation.
Collapse
Affiliation(s)
- Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
39
|
Thrikawala S, Niu M, Keller NP, Rosowski EE. Cyclooxygenase production of PGE2 promotes phagocyte control of A. fumigatus hyphal growth in larval zebrafish. PLoS Pathog 2022; 18:e1010040. [PMID: 35333905 PMCID: PMC8986117 DOI: 10.1371/journal.ppat.1010040] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/06/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
Invasive aspergillosis is a common opportunistic infection, causing >50% mortality in infected immunocompromised patients. The specific molecular mechanisms of the innate immune system that prevent pathogenesis of invasive aspergillosis in immunocompetent individuals are not fully understood. Here, we used a zebrafish larva-Aspergillus infection model to identify cyclooxygenase (COX) enzyme signaling as one mechanism that promotes host survival. Larvae exposed to the pan-COX inhibitor indomethacin succumb to infection at a significantly higher rate than control larvae. COX signaling is both macrophage- and neutrophil-mediated. However, indomethacin treatment has no effect on phagocyte recruitment. Instead, COX signaling promotes phagocyte-mediated inhibition of germination and invasive hyphal growth. Increased germination and invasive hyphal growth is also observed in infected F0 crispant larvae with mutations in genes encoding for COX enzymes (ptgs2a/b). Protective COX-mediated signaling requires the receptor EP2 and exogenous prostaglandin E2 (PGE2) rescues indomethacin-induced decreased immune control of fungal growth. Collectively, we find that COX signaling activates the PGE2-EP2 pathway to increase control A. fumigatus hyphal growth by phagocytes in zebrafish larvae.
Collapse
Affiliation(s)
- Savini Thrikawala
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Mengyao Niu
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nancy P. Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Emily E. Rosowski
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| |
Collapse
|
40
|
Wen W, Chen J, Zhou Y, Li G, Zhang Y. Loss of Ripk3 attenuated neutrophil accumulation in a lipopolysaccharide-induced zebrafish inflammatory model. Cell Death Dis 2022; 8:88. [PMID: 35220408 PMCID: PMC8882176 DOI: 10.1038/s41420-022-00891-z] [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: 12/03/2021] [Revised: 01/27/2022] [Accepted: 02/09/2022] [Indexed: 11/09/2022]
Abstract
Neutrophils are important effector cells during inflammation, which play complex roles. Therefore, investigating the regulation of neutrophil accumulation during inflammation might provide targets for treating related diseases. In the present study, we generated a ripk3-deficient zebrafish line to study the roles of Ripk3 in neutrophil-related inflammation. The homeostatic hematopoiesis and cytokine expression of the ripk3-deficient larvae were unaltered. The ripk3-deficient larvae with caudal fin fold injury exhibited similar neutrophil enrichment with wild-type larvae, suggesting that Ripk3 is not essential for non-infectious inflammatory responses. When challenged with lipopolysaccharide (LPS), the ripk3-deficient larvae showed significantly less neutrophil accumulation in the injection site and differential expression of several key cytokines. Ripk3 inhibitors could also attenuate neutrophil accumulation in wild-type larvae, indicating that Ripk3 could serve as a candidate target for inflammation treatment. In summary, our study indicated that Ripk3 has an essential role in LPS-induced inflammatory responses. It was suggested that the ripk3-deficient zebrafish might be applied in developing infectious disease models, while Ripk3 also has potential as an inflammation-treatment target.
Collapse
|
41
|
Luo K, Ogawa M, Ayer A, Britton WJ, Stocker R, Kikuchi K, Oehlers SH. Zebrafish Heme Oxygenase 1a Is Necessary for Normal Development and Macrophage Migration. Zebrafish 2022; 19:7-17. [PMID: 35108124 DOI: 10.1089/zeb.2021.0058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.
Collapse
Affiliation(s)
- Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Masahito Ogawa
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Anita Ayer
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Kazu Kikuchi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,National Cerebral and Cardiovascular Center, Suita, Japan
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Sydney Institute for Infectious Diseases, The University of Sydney, Camperdown, Australia
| |
Collapse
|
42
|
Chowdhury K, Lin S, Lai SL. Comparative Study in Zebrafish and Medaka Unravels the Mechanisms of Tissue Regeneration. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.783818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tissue regeneration has been in the spotlight of research for its fascinating nature and potential applications in human diseases. The trait of regenerative capacity occurs diversely across species and tissue contexts, while it seems to decline over evolution. Organisms with variable regenerative capacity are usually distinct in phylogeny, anatomy, and physiology. This phenomenon hinders the feasibility of studying tissue regeneration by directly comparing regenerative with non-regenerative animals, such as zebrafish (Danio rerio) and mice (Mus musculus). Medaka (Oryzias latipes) is a fish model with a complete reference genome and shares a common ancestor with zebrafish approximately 110–200 million years ago (compared to 650 million years with mice). Medaka shares similar features with zebrafish, including size, diet, organ system, gross anatomy, and living environment. However, while zebrafish regenerate almost every organ upon experimental injury, medaka shows uneven regenerative capacity. Their common and distinct biological features make them a unique platform for reciprocal analyses to understand the mechanisms of tissue regeneration. Here we summarize current knowledge about tissue regeneration in these fish models in terms of injured tissues, repairing mechanisms, available materials, and established technologies. We further highlight the concept of inter-species and inter-organ comparisons, which may reveal mechanistic insights and hint at therapeutic strategies for human diseases.
Collapse
|
43
|
Lanham KA, Nedden ML, Wise VE, Taylor MR. Genetically inducible and reversible zebrafish model of systemic inflammation. Biol Open 2022; 11:274172. [PMID: 35099005 PMCID: PMC8918989 DOI: 10.1242/bio.058559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022] Open
Abstract
The inflammatory response is a vital defense mechanism against trauma and pathogen induced damage, but equally important is its appropriate resolution. In some instances of severe trauma or sustained infection, inappropriate and persistent activation of the immune response can occur resulting in a dangerous systemic inflammatory response. Untreated, this systemic inflammatory response can lead to tissue damage, organ shutdown, and death. Replicating this condition in tractable model organisms can provide insight into the mechanisms involved in the induction, maintenance, and resolution of inflammation. To that end, we developed a non-invasive, inducible, and reversible model of systemic inflammation in zebrafish. Using the Gal4-EcR/UAS system activated by the ecdysone analog tebufenozide, we generated transgenic zebrafish that allow for chemically-induced, ubiquitous secretion of the mature form of zebrafish interleukin-1β (Il-1βmat) in both larval and adult developmental stages. To ensure a robust immune response, we attached a strong signal peptide from the Gaussia princeps luciferase enzyme to promote active secretion of the cytokine. We observe a dose-dependent inflammatory response involving neutrophil expansion accompanied by tissue damage and reduced survival. Washout of tebufenozide permits inflammation resolution. We also establish the utility of this model for the identification of small molecule anti-inflammatory compounds by treatment with the immunosuppressant rapamycin. Taken together, these features make this model a valuable new tool that can aid in identifying potential new therapies while broadening our understanding of systemic inflammation, its impact on the immune system and its resolution.
Collapse
Affiliation(s)
- Kevin A Lanham
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Megan L Nedden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Virginia E Wise
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Michael R Taylor
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
44
|
Ding L, Luo K, Feng CG, Oehlers SH. Pidotimod increases inflammation in wounded zebrafish embryos. FISH & SHELLFISH IMMUNOLOGY 2022; 120:429-433. [PMID: 34922016 DOI: 10.1016/j.fsi.2021.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Pidotimod (PDT) is a synthetic dipeptide molecule which can improve immune responses in mice and humans, protecting hosts from infection. However, the exact mechanism of protection remains ill-defined. The effect of pidotimod has not yet been investigated in the inflammatory response of zebrafish. In this study, we used tail wound and infection models of zebrafish to study the effect of PDT on inflammation. We found that zebrafish larvae were sensitive to PDT immersion causing toxicity at doses above 50 μg/mL. The tail wound assay showed that PDT increased the recruitment of neutrophils and macrophages to the wound site and promoted the transcription of the pro-inflammatory cytokine il1b. However, we did not observe protection of uropathogenic Escherichia coli or Mycobacterium marinum infected zebrafish larvae following PDT treatment. This study provides a new platform for PDT research, which is worthy of further research to identify further effects of PDT therapy.
Collapse
Affiliation(s)
- Li Ding
- Department of Infectious Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 20006, Australia; Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Carl G Feng
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 20006, Australia; Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Stefan H Oehlers
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 20006, Australia; Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
45
|
Herman A, Greenhough A, Gurevich DB. Fluorescence-Activated Cell Sorting and Quantitative Real-Time PCR to Reveal VEGF-Expressing Macrophage Populations in the Zebrafish Larvae. Methods Mol Biol 2022; 2475:325-337. [PMID: 35451769 DOI: 10.1007/978-1-0716-2217-9_24] [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] [Indexed: 06/14/2023]
Abstract
The transparent, genetically tractable zebrafish is increasingly recognized as a useful model to both live image and uncover mechanistic insight into cell interactions governing tissue homeostasis, pathology, and regeneration. Here, we describe a protocol for the isolation of macrophages from zebrafish wounds using fluorescence-activated cell sorting (FACS), and the identification of specific pro-angiogenic macrophage populations that express high levels of vascular endothelial growth factor (vegf) using quantitative real-time PCR (qPCR). The cell dissociation and FACS sorting techniques have been optimized for immune cells and successfully used to isolate other fluorescently marked populations within the wound such as neutrophils and endothelial cells. More broadly, this protocol can be easily adapted to other contexts where identification of pro-angiogenic immune cells is transformative for understanding, from development to pathologies such as infection, cancer, and diabetes.
Collapse
Affiliation(s)
- Andrew Herman
- Faculty of Life Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Alexander Greenhough
- Centre for Research in Biosciences, Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - David B Gurevich
- Department of Biology and Biochemistry, Centre for Therapeutic Innovation, Faculty of Science, University of Bath, Bath, UK.
| |
Collapse
|
46
|
Ranawat N, Masai I. Mechanisms underlying microglial colonization of developing neural retina in zebrafish. eLife 2021; 10:70550. [PMID: 34872632 PMCID: PMC8651297 DOI: 10.7554/elife.70550] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia are brain-resident macrophages that function as the first line of defense in brain. Embryonic microglial precursors originate in peripheral mesoderm and migrate into the brain during development. However, the mechanism by which they colonize the brain is incompletely understood. The retina is one of the first brain regions to accommodate microglia. In zebrafish, embryonic microglial precursors use intraocular hyaloid blood vessels as a pathway to migrate into the optic cup via the choroid fissure. Once retinal progenitor cells exit the cell cycle, microglial precursors associated with hyaloid blood vessels start to infiltrate the retina preferentially through neurogenic regions, suggesting that colonization of retinal tissue depends upon the neurogenic state. Along with blood vessels and retinal neurogenesis, IL34 also participates in microglial precursor colonization of the retina. Altogether, CSF receptor signaling, blood vessels, and neuronal differentiation function as cues to create an essential path for microglial migration into developing retina. The immune system is comprised of many different cells which protect our bodies from infection and other illnesses. The brain contains its own population of immune cells called microglia. Unlike neurons, these cells form outside the brain during development. They then travel to the brain and colonize specific regions like the retina, the light-sensing part of the eye in vertebrates. It is poorly understood how newly formed microglia migrate to the retina and whether their entry depends on the developmental state of nerve cells (also known as neurons) in this region. To help answer these questions, Ranawat and Masai attached fluorescent labels that can be seen under a microscope to microglia in the embryos of zebrafish. Developing zebrafish are transparent, making it easy to trace the fluorescent microglia as they travel to the retina and insert themselves among its neurons. Ranawat and Masai found that blood vessels around the retina act as a pathway that microglia move along. Once they reach the retina, the microglia remain attached and only enter the retina at sites where brain cells are starting to mature in to adult neurons. Further experiments showed that microglia fail to infiltrate and colonize the retina when blood vessels are damaged or neuron maturation is blocked. These findings reveal some of the key elements that guide microglia to the retina during development. However, further work is needed to establish the molecular and biochemical processes that allow microglia to attach to blood vessels and detect when cells in the retina are starting to mature.
Collapse
Affiliation(s)
- Nishtha Ranawat
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|
47
|
Ma H, Liu Z, Yang Y, Feng D, Dong Y, Garbutt TA, Hu Z, Wang L, Luan C, Cooper CD, Li Y, Welch JD, Qian L, Liu J. Functional coordination of non-myocytes plays a key role in adult zebrafish heart regeneration. EMBO Rep 2021; 22:e52901. [PMID: 34523214 DOI: 10.15252/embr.202152901] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac regeneration occurs primarily through proliferation of existing cardiomyocytes, but also involves complex interactions between distinct cardiac cell types including non-cardiomyocytes (non-CMs). However, the subpopulations, distinguishing molecular features, cellular functions, and intercellular interactions of non-CMs in heart regeneration remain largely unexplored. Using the LIGER algorithm, we assemble an atlas of cell states from 61,977 individual non-CM scRNA-seq profiles isolated at multiple time points during regeneration. This analysis reveals extensive non-CM cell diversity, including multiple macrophage (MC), fibroblast (FB), and endothelial cell (EC) subpopulations with unique spatiotemporal distributions, and suggests an important role for MC in inducing the activated FB and EC subpopulations. Indeed, pharmacological perturbation of MC function compromises the induction of the unique FB and EC subpopulations. Furthermore, we developed computational algorithm Topologizer to map the topological relationships and dynamic transitions between functional states. We uncover dynamic transitions between MC functional states and identify factors involved in mRNA processing and transcriptional regulation associated with the transition. Together, our single-cell transcriptomic analysis of non-CMs during cardiac regeneration provides a blueprint for interrogating the molecular and cellular basis of this process.
Collapse
Affiliation(s)
- Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ziqing Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yuchen Yang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dong Feng
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanhan Dong
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tiffany A Garbutt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Li Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Changfei Luan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Cynthia D Cooper
- School of Molecular Biosciences, Washington State University Vancouver, Vancouver, WA, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA.,Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
48
|
Morris S, Cholan PM, Britton WJ, Oehlers SH. Glucose inhibits haemostasis and accelerates diet-induced hyperlipidaemia in zebrafish larvae. Sci Rep 2021; 11:19049. [PMID: 34561530 PMCID: PMC8463691 DOI: 10.1038/s41598-021-98566-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
Hyperglycaemia damages the microvasculature in part through the reduced recruitment of immune cells and interference with platelet signalling, leading to poor wound healing and accelerated lipid deposition in mammals. We investigated the utility of zebrafish larvae to model the effect of exogenous glucose on neutrophil and macrophage recruitment to a tail wound, wound-induced haemostasis, and chicken egg yolk feed challenge-induced hyperlipidaemia by supplementing larvae with exogenous glucose by immersion or injection. Neither method of glucose supplementation affected the recruitment of neutrophils and macrophages following tail transection. Glucose injection reduced thrombocyte retention and fibrin plug formation while only thrombocyte retention was reduced by glucose immersion following tail transection. We observed accelerated lipid accumulation in glucose-injected larvae challenged with high fat chicken egg yolk feeding. Our study identifies conserved and divergent effects of high glucose on inflammation, haemostasis, and hyperlipidaemia in zebrafish larvae compared to mammals.
Collapse
Affiliation(s)
- Simone Morris
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Pradeep Manuneedhi Cholan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
- Discipline of Infectious Diseases and Immunology and Marie Bashir Institute, The University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
49
|
Liu S, Yang Q, Chen Y, Liu Q, Wang W, Song J, Zheng Y, Liu W. Integrated Analysis of mRNA- and miRNA-Seq in the Ovary of Rare Minnow Gobiocypris rarus in Response to 17α-Methyltestosterone. Front Genet 2021; 12:695699. [PMID: 34421998 PMCID: PMC8375321 DOI: 10.3389/fgene.2021.695699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
17α-Methyltestosterone (MT) is a synthetic androgen. The objective of this study was to explore the effects of exogenous MT on the growth and gonadal development of female rare minnow Gobiocypris rarus. Female G. rarus groups were exposed to 25–100 ng/L of MT for 7 days. After exposure for 7 days, the total weight and body length were significantly decreased in the 50-ng/L MT groups. The major oocytes in the ovaries of the control group were vitellogenic oocytes (Voc) and cortical alveolus stage oocytes (Coc). In the MT exposure groups, some fish had mature ovaries with a relatively lower proportion of mature oocytes, and the diameter of the perinucleolar oocytes (Poc) was decreased compared with those of the control group. Ovarian VTG, FSH, LH, 11-KT, E2, and T were significantly increased after exposure to 50 ng/L of MT for 7 days. Unigenes (73,449), 24 known mature microRNAs (miRNAs), and 897 novel miRNAs in the gonads of G. rarus were found using high-throughput sequencing. Six mature miRNAs (miR-19, miR-183, miR-203, miR-204, miR-205, and miR-96) as well as six differentially expressed genes (fabp3, mfap4, abca1, foxo3, tgfb1, and zfp36l1) that may be associated with ovarian development and innate immune response were assayed using qPCR. Furthermore, the miR-183 cluster and miR-203 were differentially expressed in MT-exposed ovaries of the different G. rarus groups. This study provides some information about the role of miRNA–mRNA pairs in the regulation of ovarian development and innate immune system, which will facilitate future studies of the miRNA–RNA-associated regulation of teleost reproduction.
Collapse
Affiliation(s)
- Shaozhen Liu
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Qiong Yang
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Yue Chen
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Qing Liu
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Weiwei Wang
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Jing Song
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| | - Yao Zheng
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Wenzhong Liu
- College of Animal Science, Shanxi Agriculture University, Jinzhong, China
| |
Collapse
|
50
|
Bohaud C, Johansen MD, Jorgensen C, Ipseiz N, Kremer L, Djouad F. The Role of Macrophages During Zebrafish Injury and Tissue Regeneration Under Infectious and Non-Infectious Conditions. Front Immunol 2021; 12:707824. [PMID: 34367168 PMCID: PMC8334857 DOI: 10.3389/fimmu.2021.707824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
The future of regenerative medicine relies on our understanding of the mechanistic processes that underlie tissue regeneration, highlighting the need for suitable animal models. For many years, zebrafish has been exploited as an adequate model in the field due to their very high regenerative capabilities. In this organism, regeneration of several tissues, including the caudal fin, is dependent on a robust epimorphic regenerative process, typified by the formation of a blastema, consisting of highly proliferative cells that can regenerate and completely grow the lost limb within a few days. Recent studies have also emphasized the crucial role of distinct macrophage subpopulations in tissue regeneration, contributing to the early phases of inflammation and promoting tissue repair and regeneration in late stages once inflammation is resolved. However, while most studies were conducted under non-infectious conditions, this situation does not necessarily reflect all the complexities of the interactions associated with injury often involving entry of pathogenic microorganisms. There is emerging evidence that the presence of infectious pathogens can largely influence and modulate the host immune response and the regenerative processes, which is sometimes more representative of the true complexities underlying regenerative mechanics. Herein, we present the current knowledge regarding the paths involved in the repair of non-infected and infected wounds using the zebrafish model.
Collapse
Affiliation(s)
| | - Matt D Johansen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, CHU, Montpellier, France
| | - Natacha Ipseiz
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,IRIM, INSERM, Montpellier, France
| | | |
Collapse
|