1
|
Sobah ML, Liongue C, Ward AC. Socs3a is Dispensable for Zebrafish Hematopoiesis and is Required for Neuromast Formation. FRONT BIOSCI-LANDMRK 2025; 30:36537. [PMID: 40302337 DOI: 10.31083/fbl36537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Suppressor of cytokine signaling (SOCS)3 is a regulatory protein that participates in an important negative feedback loop downstream of several critical cytokines, especially members of the interleukin-6 (IL-6) family. As a result, SOCS3 has been shown to impact the development and function of blood and immune cells. Zebrafish harbor duplicates of SOCS3, Socs3a and Socs3b, both of which possess conserved functional domains. METHODS This study explored the role of zebrafish Socs3a by creating a whole genome knockout using CRISPR/Cas9, with a focus on hematopoiesis and neuromast formation. RESULTS A zebrafish Socs3a knockout mutant was successfully generated. Characterization of this mutant revealed that normal hematopoiesis was not impacted nor was neutrophils lacking Socs3a displayed normal responses to injury or their production during emergency granulopoiesis. Neuromast formation was severely impacted in Socs3a knockout zebrafish. CONCLUSIONS Zebrafish Socs3a mutants display normal hematopoiesis and myeloid function, but the formation of the lateral line neuromast was affected by the absence of Socs3a.
Collapse
Affiliation(s)
- Mohamed Luban Sobah
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- IMPACT, Deakin University, Geelong, VIC 3220, Australia
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- IMPACT, Deakin University, Geelong, VIC 3220, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- IMPACT, Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
2
|
de Sena-Tomás C, Rebola Lameira L, Rebocho da Costa M, Naique Taborda P, Laborde A, Orger M, de Oliveira S, Saúde L. Neutrophil immune profile guides spinal cord regeneration in zebrafish. Brain Behav Immun 2024; 120:514-531. [PMID: 38925414 DOI: 10.1016/j.bbi.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/15/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Spinal cord injury triggers a strong innate inflammatory response in both non-regenerative mammals and regenerative zebrafish. Neutrophils are the first immune population to be recruited to the injury site. Yet, their role in the repair process, particularly in a regenerative context, remains largely unknown. Here, we show that, following rapid recruitment to the injured spinal cord, neutrophils mostly reverse migrate throughout the zebrafish body. In addition, promoting neutrophil inflammation resolution by inhibiting Cxcr4 boosts cellular and functional regeneration. Neutrophil-specific RNA-seq analysis reveals an enhanced activation state that correlates with a transient increase in tnf-α expression in macrophage/microglia populations. Conversely, blocking neutrophil recruitment through Cxcr1/2 inhibition diminishes the presence of macrophage/microglia at the injury site and impairs spinal cord regeneration. Altogether, these findings provide new insights into the role of neutrophils in spinal cord regeneration, emphasizing the significant impact of their immune profile on the outcome of the repair process.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Leonor Rebola Lameira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mariana Rebocho da Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Patrícia Naique Taborda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Alexandre Laborde
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Michael Orger
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - 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; Harold and Muriel Block Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Montefiore-Einstein Comprehensive Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Histologia e Biologia de Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
3
|
Hauser KA, Garvey CN, Crow RS, Hossainey MRH, Howard DT, Ranganathan N, Gentry LK, Yaparla A, Kalia N, Zelle M, Jones EJ, Duttargi AN, Rollins-Smith LA, Muletz-Wolz CR, Grayfer L. Amphibian mast cells serve as barriers to chytrid fungus infections. eLife 2024; 12:RP92168. [PMID: 39082933 PMCID: PMC11290838 DOI: 10.7554/elife.92168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Global amphibian declines are compounded by deadly disease outbreaks caused by the chytrid fungus, Batrachochytrium dendrobatidis (Bd). Much has been learned about the roles of amphibian skin-produced antimicrobial components and microbiomes in controlling Bd, yet almost nothing is known about the roles of skin-resident immune cells in anti-Bd defenses. Mammalian mast cells reside within and serve as key immune sentinels in barrier tissues like skin. Accordingly, we investigated the roles of Xenopus laevis frog mast cells during Bd infections. Our findings indicate that enrichment of X. laevis skin mast cells confers anti-Bd protection and ameliorates the inflammation-associated skin damage caused by Bd infection. This includes a significant reduction in infiltration of Bd-infected skin by neutrophils, promoting mucin content within cutaneous mucus glands, and preventing Bd-mediated changes to skin microbiomes. Mammalian mast cells are known for their production of the pleiotropic interleukin-4 (IL4) cytokine and our findings suggest that the X. laevis IL4 plays a key role in manifesting the effects seen following cutaneous mast cell enrichment. Together, this work underscores the importance of amphibian skin-resident immune cells in anti-Bd defenses and illuminates a novel avenue for investigating amphibian host-chytrid pathogen interactions.
Collapse
Affiliation(s)
- Kelsey A Hauser
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Christina N Garvey
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Ryley S Crow
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Muhammad RH Hossainey
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Dustin T Howard
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Netra Ranganathan
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Lindsey K Gentry
- Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology InstituteWashingtonUnited States
| | - Amulya Yaparla
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Namarta Kalia
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Mira Zelle
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| | - Elizabeth J Jones
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown UniversityWashingtonUnited States
| | - Anju N Duttargi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown UniversityWashingtonUnited States
| | - Louise A Rollins-Smith
- Departments of Pathology, Microbiology and Immunology, and of Pediatrics, Vanderbilt University School of MedicineNashvilleUnited States
- Department of Biological Sciences, Vanderbilt UniversityNashvilleUnited States
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian National Zoo & Conservation Biology InstituteWashingtonUnited States
| | - Leon Grayfer
- Department of Biological Sciences, The George Washington UniversityWashingtonUnited States
| |
Collapse
|
4
|
Hoyberghs J, Coppens A, Bars C, Van Ginneken C, Foubert K, Van Cruchten S. Assessing developmental toxicity and non-CYP mediated biotransformation of two anti-epileptics and their human metabolites in zebrafish embryos and larvae. Curr Res Toxicol 2024; 7:100186. [PMID: 39188273 PMCID: PMC11347070 DOI: 10.1016/j.crtox.2024.100186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/28/2024] [Accepted: 07/09/2024] [Indexed: 08/28/2024] Open
Abstract
Zebrafish embryo-based assays are a promising alternative for animal testing to screen new compounds for developmental toxicity. However, recent studies in zebrafish embryos showed an immature intrinsic cytochrome P450 (CYP)-mediated biotransformation capacity, as most CYPs were only active at the end of the organogenesis period. Data on other phase I enzymes involved in the biotransformation of xenobiotics in zebrafish embryos is limited. This information is pivotal for proteratogens needing bioactivation to exert their teratogenic potential. Therefore, this study aimed to investigate whether carbamazepine (CBZ) and levetiracetam (LTC), two anti-epileptic drugs that require bioactivation to exert their teratogenic potential, are biotransformed into non-CYP mediated metabolites in the zebrafish embryo and whether one or more of these metabolites cause developmental toxicity in this species. In the first step, zebrafish embryos were exposed to LTC and CBZ and their non-CYP mediated human metabolites, etiracetam carboxylic acid (ECA) and 9-acridine carboxaldehyde (9ACA), acridine (AI), and acridone (AO), respectively, from 5.25 to 120 hpf and morphologically evaluated. Next, the uptake of all compounds and the formation of the metabolites were assessed using LC-MS methods. As LTC and ECA were, respectively, poorly or not taken up by zebrafish larvae during the exposure experiments, we could not determine if LTC and ECA are teratogenic. However, biotransformation of LTC into ECA was observed at 24 hpf and 120 hpf, which indicates that the special type of B-esterase is already active at 24 hpf. CBZ and its three metabolites were teratogenic, as a significant increase in malformed embryos was observed for all of them. All three metabolites were more potent teratogens than CBZ, with AI being the most potent, followed by 9ACA and AO. The myeloperoxidase (MPO) homologue is already active at 24 hpf, as CBZ was biotransformed into 9ACA and AO in 24 hpf zebrafish embryos, and into 9ACA in 120 hpf larvae. Moreover, 9ACA was also found to be biotransformed into AI and AO, and AI into AO. As such, one or more of these metabolites probably contribute to the teratogenic effects observed in zebrafish larvae after exposure to CBZ.
Collapse
Affiliation(s)
- Jente Hoyberghs
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Axelle Coppens
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Chloé Bars
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Chris Van Ginneken
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Kenn Foubert
- Natural Products & Food Research and Analysis-Pharmaceutical Technology (NatuRAPT), Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Comparative Perinatal Development, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
5
|
Maéno M, Tanabe M, Ogawa A, Kobayashi H, Izutsu Y, Kato T. Identification and characterization of myeloid cells localized in the tadpole liver cortex in Xenopus laevis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105178. [PMID: 38599553 DOI: 10.1016/j.dci.2024.105178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
In the present study, using transgenic frogs that express GFP specifically in myeloid cells under the myeloperoxidase enhancer sequence, we found that myeloperoxidase-positive cells are localized in the liver cortex at the late tadpole stages. Immunohistochemical analysis revealed that myelopoiesis in the liver cortex became evident after st. 50 and reached its peak by st. 56. Transplantation experiments indicated that cells with a high density at the liver cortex were derived from the dorso-lateral plate tissue in the neurula embryo. Analysis of smear samples of the cells isolated from collagenase-treated liver tissues of the transgenic tadpoles indicated that myeloid cells were the major population of blood cells in the larval liver and that, in addition to myeloid colonies, erythroid colonies expanded in entire liver after metamorphosis. Cells that were purified from the livers of transgenic tadpoles according to the GFP expression exhibited the multi-lobed nuclei. The results of present study provide evidence that the liver cortex of the Xenopus tadpole is a major site of granulopoiesis.
Collapse
Affiliation(s)
- Mitsugu Maéno
- Institute of Science and Technology, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata, 950-2181, Japan.
| | - Miki Tanabe
- Faculty of Science, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata, 950-2181, Japan
| | - Ayame Ogawa
- Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns building, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Department of Biology, School of Education, Waseda University, Center for Advanced Biomedical Science, TWIns building, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Haruka Kobayashi
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata, 950-2181, Japan
| | - Yumi Izutsu
- Institute of Science and Technology, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata, 950-2181, Japan
| | - Takashi Kato
- Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns building, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Department of Biology, School of Education, Waseda University, Center for Advanced Biomedical Science, TWIns building, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| |
Collapse
|
6
|
Li H, Liu H, Bi L, Liu Y, Jin L, Peng R. Immunotoxicity of microplastics in fish. FISH & SHELLFISH IMMUNOLOGY 2024; 150:109619. [PMID: 38735599 DOI: 10.1016/j.fsi.2024.109619] [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: 01/09/2024] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Plastic waste degrades slowly in aquatic environments, transforming into microplastics (MPs) and nanoplastics (NPs), which are subsequently ingested by fish and other aquatic organisms, causing both physical blockages and chemical toxicity. The fish immune system serves as a crucial defense against viruses and pollutants present in water. It is imperative to comprehend the detrimental effects of MPs on the fish immune system and conduct further research on immunological assessments. In this paper, the immune response and immunotoxicity of MPs and its combination with environmental pollutants on fish were reviewed. MPs not only inflict physical harm on the natural defense barriers like fish gills and vital immune organs such as the liver and intestinal tract but also penetrate cells, disrupting intracellular signaling pathways, altering the levels of immune cytokines and gene expression, perturbing immune homeostasis, and ultimately compromising specific immunity. Initially, fish exposed to MPs recruit a significant number of macrophages and T cells while activating lysosomes. Over time, this exposure leads to apoptosis of immune cells, a decline in lysosomal degradation capacity, lysosomal activity, and complement levels. MPs possess a small specific surface area and can efficiently bind with heavy metals, organic pollutants, and viruses, enhancing immune responses. Hence, there is a need for comprehensive studies on the shape, size, additives released from MPs, along with their immunotoxic effects and mechanisms in conjunction with other pollutants and viruses. These studies aim to solidify existing knowledge and delineate future research directions concerning the immunotoxicity of MPs on fish, which has implications for human health.
Collapse
Affiliation(s)
- Huiqi Li
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Huanpeng Liu
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Liuliu Bi
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yinai Liu
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Libo Jin
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Renyi Peng
- Affiliation: Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
7
|
Hammond FR, Lewis A, Pollara G, Tomlinson GS, Noursadeghi M, Kiss-Toth E, Elks PM. Tribbles1 is host protective during in vivo mycobacterial infection. eLife 2024; 13:e95980. [PMID: 38896446 PMCID: PMC11186633 DOI: 10.7554/elife.95980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1β and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.
Collapse
Affiliation(s)
- Ffion R Hammond
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Amy Lewis
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Gabriele Pollara
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Gillian S Tomlinson
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Mahdad Noursadeghi
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Endre Kiss-Toth
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
8
|
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] [Grants] [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
|
9
|
Sobah ML, Liongue C, Ward AC. Stat3 Regulates Developmental Hematopoiesis and Impacts Myeloid Cell Function via Canonical and Non-Canonical Modalities. J Innate Immun 2024; 16:262-282. [PMID: 38643762 PMCID: PMC11249464 DOI: 10.1159/000538364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/12/2024] [Indexed: 04/23/2024] Open
Abstract
INTRODUCTION Signal transducer and activator of transcription (STAT) 3 is extensively involved in the development, homeostasis, and function of immune cells, with STAT3 disruption associated with human immune-related disorders. The roles ascribed to STAT3 have been assumed to be due to its canonical mode of action as an inducible transcription factor downstream of multiple cytokines, although alternative noncanonical functional modalities have also been identified. The relative involvement of each mode was further explored in relevant zebrafish models. METHODS Genome editing with CRISPR/Cas9 was used to generate mutants of the conserved zebrafish Stat3 protein: a loss of function knockout (KO) mutant and a mutant lacking C-terminal sequences including the transactivation domain (ΔTAD). Lines harboring these mutations were analyzed with respect to blood and immune cell development and function in comparison to wild-type zebrafish. RESULTS The Stat3 KO mutant showed perturbation of hematopoietic lineages throughout primitive and early definitive hematopoiesis. Neutrophil numbers did not increase in response to lipopolysaccharide (LPS) or granulocyte colony-stimulating factor (G-CSF) and their migration was significantly diminished, the latter correlating with abrogation of the Cxcl8b/Cxcr2 pathway, with macrophage responses perturbed. Intriguingly, many of these phenotypes were not shared by the Stat3 ΔTAD mutant. Indeed, only neutrophil and macrophage development were disrupted in these mutants with responsiveness to LPS and G-CSF maintained, and neutrophil migration actually increased. CONCLUSION This study has identified roles for zebrafish Stat3 within hematopoietic stem cells impacting multiple lineages throughout primitive and early definitive hematopoiesis, myeloid cell responses to G-CSF and LPS and neutrophil migration. Many of these roles showed conservation, but notably several involved noncanonical modalities, providing additional insights for relevant diseases.
Collapse
Affiliation(s)
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| |
Collapse
|
10
|
Kirchberger S, Shoeb MR, Lazic D, Wenninger-Weinzierl A, Fischer K, Shaw LE, Nogueira F, Rifatbegovic F, Bozsaky E, Ladenstein R, Bodenmiller B, Lion T, Traver D, Farlik M, Schöfer C, Taschner-Mandl S, Halbritter F, Distel M. Comparative transcriptomics coupled to developmental grading via transgenic zebrafish reporter strains identifies conserved features in neutrophil maturation. Nat Commun 2024; 15:1792. [PMID: 38413586 PMCID: PMC10899643 DOI: 10.1038/s41467-024-45802-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Neutrophils are evolutionarily conserved innate immune cells playing pivotal roles in host defense. Zebrafish models have contributed substantially to our understanding of neutrophil functions but similarities to human neutrophil maturation have not been systematically characterized, which limits their applicability to studying human disease. Here we show, by generating and analysing transgenic zebrafish strains representing distinct neutrophil differentiation stages, a high-resolution transcriptional profile of neutrophil maturation. We link gene expression at each stage to characteristic transcription factors, including C/ebp-β, which is important for late neutrophil maturation. Cross-species comparison of zebrafish, mouse, and human samples confirms high molecular similarity of immature stages and discriminates zebrafish-specific from pan-species gene signatures. Applying the pan-species neutrophil maturation signature to RNA-sequencing data from human neuroblastoma patients reveals association between metastatic tumor cell infiltration in the bone marrow and an overall increase in mature neutrophils. Our detailed neutrophil maturation atlas thus provides a valuable resource for studying neutrophil function at different stages across species in health and disease.
Collapse
Grants
- I 4162 Austrian Science Fund FWF
- TAI 454 Austrian Science Fund FWF
- TAI 732 Austrian Science Fund FWF
- St. Anna Kinderkrebsforschung (to S.T.M., R.L., F.H., and M.D.), the Austrian Research Promotion Agency (FFG) (project 7940628, Danio4Can to M.D.), a German Academic Exchange Service postdoctoral fellowship and an EMBO fellowship (to M.D.), the Austrian Science Fund (FWF) through grants TAI454 (to F.H. and M.D.), TAI732 (to F.H.), I4162 (ERA-NET/Transcan-2 LIQUIDHOPE; to S.T.M.), P35841 (MAPMET; to S.T.M.), P34152 (to T.L.), P 30642 (to C.S.) and the Alex’s Lemonade Stand Foundation for Childhood Cancer 20-17258 (to F.H. and M.D.), and the Swiss Government Excellence Scholarship (to D.L.), and the EC H2020 grant no. 826494 (PRIMAGE; to R.L.), and by the European Commission within the FP7 Framework program (Fungitect-Grant No 602125 to T.L.).
Collapse
Affiliation(s)
| | - Mohamed R Shoeb
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Daria Lazic
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | | | - Kristin Fischer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Lisa E Shaw
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Filomena Nogueira
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia - Labordiagnostik GmbH, Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs, Campus Vienna Biocenter, Vienna, Austria
| | | | - Eva Bozsaky
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Ruth Ladenstein
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Thomas Lion
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia - Labordiagnostik GmbH, Vienna, Austria
- Medical University of Vienna, Department of Pediatrics, Vienna, Austria
| | - David Traver
- Cell and Developmental Biology, University of California, San Diego, CA, USA
| | - Matthias Farlik
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Christian Schöfer
- Medical University of Vienna, Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Vienna, Austria
| | | | | | - Martin Distel
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
| |
Collapse
|
11
|
Sobah ML, Liongue C, Ward AC. Contribution of Signal Transducer and Activator of Transcription 3 (STAT3) to Bone Development and Repair. Int J Mol Sci 2023; 25:389. [PMID: 38203559 PMCID: PMC10778865 DOI: 10.3390/ijms25010389] [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: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor activated canonically by numerous cytokines and other factors, with significant roles in immunity, immune diseases, and cancer. It has also been implicated in several human skeletal disorders, with loss-of-function (LOF) mutations associated with aberrant skeletal development. To gain further insights, two zebrafish STAT3 lines were investigated: a complete LOF knockout (KO) mutant and a partial LOF mutant with the transactivation domain truncated (ΔTAD). Consistent with other studies, the KO mutants were smaller, with reduced length in early embryos exacerbated by a decreased growth rate from 5 days postfertilization (dpf). They displayed skeletal deformities that approached 80% incidence by 30 dpf, with a significant reduction in early bone but not cartilage formation. Further analysis additionally identified considerable abrogation of caudal fin regeneration, concomitant with a paucity of infiltrating macrophages and neutrophils, which may be responsible for this. Most of these phenotypes were also observed in the ΔTAD mutants, indicating that loss of canonical STAT3 signaling was the likely cause. However, the impacts on early bone formation and regeneration were muted in the ΔTAD mutant, suggesting the potential involvement of noncanonical functions in these processes.
Collapse
Affiliation(s)
- Mohamed L. Sobah
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Clifford Liongue
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Alister C. Ward
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| |
Collapse
|
12
|
Khanjani MH, Sharifinia M, Emerenciano MGC. A detailed look at the impacts of biofloc on immunological and hematological parameters and improving resistance to diseases. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108796. [PMID: 37149233 DOI: 10.1016/j.fsi.2023.108796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
The innate immunity of invertebrates serves as a critical trait that provides a valuable foundation for studying the common biological responses to environmental changes. With the exponential growth of the human population, the demand for protein has soared, leading to the intensification of aquaculture. Regrettably, this intensification has resulted in the overuse of antibiotics and chemotherapeutics, which have led to the emergence of resistant microbes or superbugs. In this regard, biofloc technology (BFT) emerges as a promising strategy for disease management in aquaculture. By harnessing the power of antibiotics, probiotics, and prebiotics, BFT offers a sustainable and eco-friendly approach that can help mitigate the negative impacts of harmful chemicals. By adopting this innovative technology, we can enhance the immunity and promote the health of aquatic organisms, thereby ensuring the long-term viability of the aquaculture industry. Using a proper carbon to nitrogen ratio, normally adding an external carbon source, BFT recycles waste in culture system with no water exchange. Heterotrophic bacteria grow along with other key microbes in the culture water. Heterotrophs play a major role in assimilating ammonia from feed and fecal waste, crucial pathway to form suspended microbial aggregates (known as 'biofloc'); while chemoautotrophs (e.g. nitrifying bacteria) oxidize ammonia into nitrite, and nitrite into nitrate promoting a healthy farming conditions. By using a highly aerated media and an organic substrates that contain carbon and nitrogen, protein-rich microbes are able to flocculate in culture water. Several types of microorganisms and their cell components have been studied and applied to aquatic animals as probiotics or immunostimulants (lipopolysaccharide, peptidoglycan, and 1-glucans) to enhance their innate immunity and antioxidant status, thereby enhancing their resistance to disease. In recent years, many studies have been conducted on the application of BFT for different farmed aquatic species and it has been observed as a promising method for the development of sustainable aquaculture, especially due to less use of water, increased productivity and biosecurity, but also an enhancement of the health status of several aquaculture species. This review analyses the immune status, antioxidant activity, blood and biochemical parameters, and level of resistance against pathogenic agents of aquatic animals farmed in BFT systems. This manuscript aims to gather and showcase the scientific evidences related to biofloc as a 'health promoter' in a unique document for the industry and academia.
Collapse
Affiliation(s)
- Mohammad Hossein Khanjani
- Department of Fisheries Sciences and Engineering, Faculty of Natural Resources, University of Jiroft, Jiroft, Kerman, Iran.
| | - Moslem Sharifinia
- Shrimp Research Center, Iranian Fisheries Sciences Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Bushehr, 75169-89177, Iran.
| | | |
Collapse
|
13
|
Biofloc Technology in Fish Aquaculture: A Review. Antioxidants (Basel) 2023; 12:antiox12020398. [PMID: 36829957 PMCID: PMC9952110 DOI: 10.3390/antiox12020398] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
The application of biofloc to fish species has several advantages, including the enhancement of production by increasing growth performance and survival rate and the improvement of fish aquaculture physiological activity. There has been a recent increase in biofloc addition to fish culture, and this review examines changes this causes to the survival and growth rate of fish and its economic feasibility. Physiological activity and disease resistance of biofloc-fed fish is being extensively studied. The hematological parameters and antioxidant and immune responses of fish fed biofloc were reviewed in this study, as well as their disease resistance by testing them for major specific diseases. Standards for effectively applying biofloc to fish aquaculture are also suggested.
Collapse
|
14
|
Sobah ML, Scott AC, Laird M, Koole C, Liongue C, Ward AC. Socs3b regulates the development and function of innate immune cells in zebrafish. Front Immunol 2023; 14:1119727. [PMID: 36969252 PMCID: PMC10030509 DOI: 10.3389/fimmu.2023.1119727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Suppressor of cytokine signaling 3 (SOCS3) is a critical component of the negative feedback regulation that controls signaling by cytokines and other factors thereby ensuring that important processes such as hematopoiesis and inflammation occur at appropriate levels. Methods To gain further insights into SOCS3 function, the zebrafish socs3b gene was investigated through analysis of a knockout line generated using CRISPR/Cas9-mediated genome editing. Results Zebrafish socs3b knockout embryos displayed elevated numbers of neutrophils during primitive and definitive hematopoiesis but macrophage numbers were not altered. However, the absence of socs3b reduced neutrophil functionality but enhanced macrophage responses. Adult socs3b knockout zebrafish displayed reduced survival that correlated with an eye pathology involving extensive infiltration of neutrophils and macrophages along with immune cell dysregulation in other tissues. Discussion These findings identify a conserved role for Socs3b in the regulation of neutrophil production and macrophage activation.
Collapse
Affiliation(s)
| | - Aimee C. Scott
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Miranda Laird
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Cassandra Koole
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
- *Correspondence: Alister C. Ward,
| |
Collapse
|
15
|
Shanaka KASN, Jung S, Madushani KP, Wijerathna HMSM, Neranjan Tharuka MD, Kim MJ, Lee J. Generation of viperin-knockout zebrafish by CRISPR/Cas9-mediated genome engineering and the effect of this mutation under VHSV infection. FISH & SHELLFISH IMMUNOLOGY 2022; 131:672-681. [PMID: 36309322 DOI: 10.1016/j.fsi.2022.10.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
Viperin is an important virus-induced protein in animals that negatively participates in RNA viral replication and transcription. The reactive machinery of viperin suggests that it produces a regulatory molecule ddhCTP, which may affect immune regulation. In this study, we investigated the expression pattern of viperin in larval and adult stages of zebrafish by whole-mount in situ hybridization and reverse transcription-quantitative PCR (RT-qPCR). To elucidate the function of viperin, we generated a zebrafish knockout model using the CRISPR/Cas9 method and evaluated the mutation's effects under viral hemorrhagic septicemia virus (VHSV) infections. In zebrafish larvae, viperin was expressed in the brain region, eye, and pharynx, which was confirmed by cryosectioning. In adult zebrafish, blood cells showed the highest levels of viperin expression. In 5 dpf fish challenged with VHSV, the expression of the viral NP protein was significantly enhanced in viperin-/- compared to wild-type fish. In vitro VHSV propagation analysis indicated comparatively higher levels of virus propagation in viperin-/- fish. Mortality analysis confirmed higher mortality rates, and interferon gene expression analysis showed a strong upregulation of interferon (ifn)φ1 and 3 gene in viperin-/- fish infected with VHSV. This study describes the successful generation of a viperin-knockout model and the role of viperin during VHSV infections.
Collapse
Affiliation(s)
- K A S N Shanaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - K P Madushani
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - H M S M Wijerathna
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - M D Neranjan Tharuka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Myoung-Jin Kim
- Nakdonggang National Institute of Biological Resources, Sangju-si, Gyeongsangbuk-do, 37242, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self, Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
16
|
Sanguinarine Enhances the Integrity of the Blood-Milk Barrier and Inhibits Oxidative Stress in Lipopolysaccharide-Stimulated Mastitis. Cells 2022; 11:cells11223658. [PMID: 36429086 PMCID: PMC9688596 DOI: 10.3390/cells11223658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Mastitis is a common clinical disease which threatens the welfare and health of dairy cows and causes huge economic losses. Sanguinarine (SG) is a plant-derived alkaloid which has many biological functions, including antibacterial and antioxidant properties. The present study attempted to evaluate the effect of SG on lipopolysaccharide (LPS)-induced oxidative stress reactions and explore its potential mechanisms. The expression profile of SG was analyzed by network pharmacology, and it was found that differentially expressed genes were mainly involved in the Wnt signaling pathway and oxidative stress through GO and KEGG enrichment. In in vitro experiments, the dosage of SG was non-toxic to mouse mammary epithelial cells (mMECs) (p > 0.05). SG not only inhibited the increase in ROS induced by LPS, but also enhanced the activity of antioxidant enzymes (p < 0.05). Moreover, the results of the in vivo experiments showed that SG alleviated LPS-induced inflammatory damage of mouse mammary glands and enhanced the integrity of the blood-milk barrier (p < 0.05). Further studies suggested that SG promoted Nrf2 expression and suppressed the activation of the Wnt signaling pathway (p < 0.05). Conclusively, this study clarified the protective effect of SG on mastitis and provided evidence for new potential mechanisms. SG exerted its antioxidant function through activating Nrf2 and inhibiting the Wnt/β-catenin pathway, repairing the blood-milk barrier.
Collapse
|
17
|
Charlie-Silva I, Feitosa NM, Pontes LG, Fernandes BH, Nóbrega RH, Gomes JMM, Prata MNL, Ferraris FK, Melo DC, Conde G, Rodrigues LF, Aracati MF, Corrêa-Junior JD, Manrique WG, Superio J, Garcez AS, Conceição K, Yoshimura TM, Núñez SC, Eto SF, Fernandes DC, Freitas AZ, Ribeiro MS, Nedoluzhko A, Lopes-Ferreira M, Borra RC, Barcellos LJG, Perez AC, Malafaia G, Cunha TM, Belo MAA, Galindo-Villegas J. Plasma proteome responses in zebrafish following λ-carrageenan-Induced inflammation are mediated by PMN leukocytes and correlate highly with their human counterparts. Front Immunol 2022; 13:1019201. [PMID: 36248846 PMCID: PMC9559376 DOI: 10.3389/fimmu.2022.1019201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022] Open
Abstract
Regulation of inflammation is a critical process for maintaining physiological homeostasis. The λ-carrageenan (λ-CGN) is a mucopolysaccharide extracted from the cell wall of red algae (Chondrus crispus) capable of inducing acute intestinal inflammation, which is translated into the production of acute phase reactants secreted into the blood circulation. However, the associated mechanisms in vertebrates are not well understood. Here, we investigated the crucial factors behind the inflammatory milieu of λ-CGN-mediated inflammation administered at 0, 1.75, and 3.5% (v/w) by i.p. injection into the peritoneal cavity of adult zebrafish (ZF) (Danio rerio). We found that polymorphonuclear leukocytes (neutrophils) and lymphocytes infiltrating the ZF peritoneal cavity had short-term persistence. Nevertheless, they generate a strong pattern of inflammation that affects systemically and is enough to produce edema in the cavity. Consistent with these findings, cell infiltration, which causes notable tissue changes, resulted in the overexpression of several acute inflammatory markers at the protein level. Using reversed-phase high-performance liquid chromatography followed by a hybrid linear ion-trap mass spectrometry shotgun proteomic approach, we identified 2938 plasma proteins among the animals injected with PBS and 3.5% λ-CGN. First, the bioinformatic analysis revealed the composition of the plasma proteome. Interestingly, 72 commonly expressed proteins were recorded among the treated and control groups, but, surprisingly, 2830 novel proteins were differentially expressed exclusively in the λ-CGN-induced group. Furthermore, from the commonly expressed proteins, compared to the control group 62 proteins got a significant (p < 0.05) upregulation in the λ-CGN-treated group, while the remaining ten proteins were downregulated. Next, we obtained the major protein-protein interaction networks between hub protein clusters in the blood plasma of the λ-CGN induced group. Moreover, to understand the molecular underpinnings of these effects based on the unveiled protein sets, we performed a bioinformatic structural similarity analysis and generated overlapping 3D reconstructions between ZF and humans during acute inflammation. Biological pathway analysis pointed to the activation and abundance of diverse classical immune and acute phase reactants, several catalytic enzymes, and varied proteins supporting the immune response. Together, this information can be used for testing and finding novel pharmacological targets to treat human intestinal inflammatory diseases.
Collapse
Affiliation(s)
| | - Natália M. Feitosa
- Integrated Laboratory of Translational Bioscience, Institute of Biodiversity and Sustainability, Federal University of Rio de Janeiro, Macaé, Brazil
| | | | - Bianca H. Fernandes
- Laboratório de Controle Genético e Sanitário, Faculdade de Medicina Universidade de São Paulo, São Paulo, Brazil
| | - Rafael H. Nóbrega
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, São Paulo State University, São Paulo, Brazil
| | - Juliana M. M. Gomes
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Mariana N. L. Prata
- Department of Pharmacology, Instituto de CiênciasBiomédicas-Universidade Federal de Minas Gerais (ICB-UFMG), Belo Horizonte, Brazil
| | - Fausto K. Ferraris
- Department of Pharmacology and Toxicology, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Daniela C. Melo
- Laboratory of Zebrafish from Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gabriel Conde
- Department of Preventive Veterinary Medicine, São Paulo State University, São Paulo, Brazil
| | - Letícia F. Rodrigues
- Department of Preventive Veterinary Medicine, São Paulo State University, São Paulo, Brazil
| | - Mayumi F. Aracati
- Department of Preventive Veterinary Medicine, São Paulo State University, São Paulo, Brazil
| | - José D. Corrêa-Junior
- Department of Morphology, Instituto de CiênciasBiomédicas-Universidade Federal de Minas Gerais (ICB-UFMG), Belo Horizonte, Brazil
| | - Wilson G. Manrique
- Veterinary College, Federal University of Rondonia, Rolim de Moura, Brazil
| | - Joshua Superio
- Department of Aquaculture, Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Katia Conceição
- Peptide Biochemistry Laboratory, Universidade Federal de São Paulo (UNIFESP), Sao Jose Dos Campos, Brazil
| | - Tania M. Yoshimura
- Center for Lasers and Applications, Instituto de PesquisasEnergéticas e Nucleares (IPEN-CNEN), Sao Paulo, Brazil
| | - Silvia C. Núñez
- University Brazil, São Paulo, Brazil
- University Brazil, Descalvado, Brazil
| | - Silas F. Eto
- Development and Innovation Laboratory, Center of Innovation and Development, Butantan Institute, São Paulo, Brazil
| | - Dayanne C. Fernandes
- Department of Preventive Veterinary Medicine, São Paulo State University, São Paulo, Brazil
| | - Anderson Z. Freitas
- Center for Lasers and Applications, Instituto de PesquisasEnergéticas e Nucleares (IPEN-CNEN), Sao Paulo, Brazil
| | - Martha S. Ribeiro
- Center for Lasers and Applications, Instituto de PesquisasEnergéticas e Nucleares (IPEN-CNEN), Sao Paulo, Brazil
| | - Artem Nedoluzhko
- Paleogenomics Laboratory, European University at Saint Petersburg, Saint Petersburg, Russia
| | | | - Ricardo C. Borra
- Department of Genetics and Evolution, Federal University of São Carlos, São Paulo, Brazil
| | - Leonardo J. G. Barcellos
- Postgraduate Program in Pharmacology, Federal University of Santa Maria, Rio Grande do Sul, Brazil
- Postgraduate Program in Bioexperimentation. University of Passo Fundo, Rio Grande do Sul, Brazil
| | - Andrea C. Perez
- Department of Pharmacology and Toxicology, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Guilheme Malafaia
- Biological Research Laboratory, Goiano Federal Institute, Urutaí, Brazil
| | - Thiago M. Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Marco A. A. Belo
- Department of Preventive Veterinary Medicine, São Paulo State University, São Paulo, Brazil
- University Brazil, São Paulo, Brazil
- University Brazil, Descalvado, Brazil
| | - Jorge Galindo-Villegas
- Department of Genomics, Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
18
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
19
|
Myllymäki H, Yu PP, Feng Y. Opportunities presented by zebrafish larval models to study neutrophil function in tissues. Int J Biochem Cell Biol 2022; 148:106234. [PMID: 35667555 DOI: 10.1016/j.biocel.2022.106234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 10/18/2022]
Abstract
Appropriate neutrophil function is essential for innate immune defence and to avoid inflammatory pathology. Neutrophils can adapt their responses according to their environment and recently, the existence of multiple distinct neutrophil populations has been confirmed in both health and disease. However, the study of neutrophil functions in their tissue environment has remained challenging, and for instance, the relationship between neutrophil maturity and function is not fully understood. Many neutrophil morphological and functional features are highly conserved between mammals and non-mammalian vertebrates. This enables the use of the transparent and genetically tractable zebrafish larvae to study neutrophil biology. We review data on the development and function of zebrafish larval neutrophils and advances zebrafish have brought to studies of neutrophil biology. In addition, we discuss opportunities and aspects to be considered when using the larval zebrafish model to further enhance our understanding of neutrophil function in health and disease.
Collapse
Affiliation(s)
- Henna Myllymäki
- UoE Centre for Inflammation Research, Queen's Medical Research Institute, Institute for Regeneration and Repair, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Peiyi Pearl Yu
- UoE Centre for Inflammation Research, Queen's Medical Research Institute, Institute for Regeneration and Repair, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Yi Feng
- UoE Centre for Inflammation Research, Queen's Medical Research Institute, Institute for Regeneration and Repair, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
20
|
Otterstrom JJ, Lubin A, Payne EM, Paran Y. Technologies bringing young Zebrafish from a niche field to the limelight. SLAS Technol 2022; 27:109-120. [PMID: 35058207 DOI: 10.1016/j.slast.2021.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Fundamental life science and pharmaceutical research are continually striving to provide physiologically relevant context for their biological studies. Zebrafish present an opportunity for high-content screening (HCS) to bring a true in vivo model system to screening studies. Zebrafish embryos and young larvae are an economical, human-relevant model organism that are amenable to both genetic engineering and modification, and direct inspection via microscopy. The use of these organisms entails unique challenges that new technologies are overcoming, including artificial intelligence (AI). In this perspective article, we describe the state-of-the-art in terms of automated sample handling, imaging, and data analysis with zebrafish during early developmental stages. We highlight advances in orienting the embryos, including the use of robots, microfluidics, and creative multi-well plate solutions. Analyzing the micrographs in a fast, reliable fashion that maintains the anatomical context of the fluorescently labeled cells is a crucial step. Existing software solutions range from AI-driven commercial solutions to bespoke analysis algorithms. Deep learning appears to be a critical tool that researchers are only beginning to apply, but already facilitates many automated steps in the experimental workflow. Currently, such work has permitted the cellular quantification of multiple cell types in vivo, including stem cell responses to stress and drugs, neuronal myelination and macrophage behavior during inflammation and infection. We evaluate pro and cons of proprietary versus open-source methodologies for combining technologies into fully automated workflows of zebrafish studies. Zebrafish are poised to charge into HCS with ever-greater presence, bringing a new level of physiological context.
Collapse
Affiliation(s)
| | - Alexandra Lubin
- Research Department of Hematology, Cancer Institute, University College London, London, UK
| | - Elspeth M Payne
- Research Department of Hematology, Cancer Institute, University College London, London, UK
| | | |
Collapse
|
21
|
Zang L, Torraca V, Shimada Y, Nishimura N. Editorial: Zebrafish Models for Human Disease Studies. Front Cell Dev Biol 2022; 10:861941. [PMID: 35359457 PMCID: PMC8963268 DOI: 10.3389/fcell.2022.861941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Liqing Zang
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
- *Correspondence: Liqing Zang, ; Vincenzo Torraca, ; Yasuhito Shimada,
| | - Vincenzo Torraca
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
- School of Life Sciences, University of Westminster, London, United Kingdom
- *Correspondence: Liqing Zang, ; Vincenzo Torraca, ; Yasuhito Shimada,
| | - Yasuhito Shimada
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
- *Correspondence: Liqing Zang, ; Vincenzo Torraca, ; Yasuhito Shimada,
| | - Norihiro Nishimura
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| |
Collapse
|
22
|
Cheng H, Duan Z, Wu Y, Wang Y, Zhang H, Shi Y, Zhang H, Wei Y, Sun H. Immunotoxicity responses to polystyrene nanoplastics and their related mechanisms in the liver of zebrafish (Danio rerio) larvae. ENVIRONMENT INTERNATIONAL 2022; 161:107128. [PMID: 35134711 DOI: 10.1016/j.envint.2022.107128] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/18/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Nanoplastics in aquatic environments may induce adverse immunotoxicity effects in fish. However, there is insufficient evidence on the visible immunotoxicity endpoints in the larval stages of fish. The liver plays an important role in systemic and local innate immunity in the fish. In this study, the hepatic inflammatory effects of polystyrene (PS) nanoplastic particles (NPs: 100 and 50 nm) and micron PS particles on transgenic zebrafish (Danio rerio) larvae were estimated using fluorescent-labeled neutrophils, macrophages, and liver-type inflammatory binding protein (fabp10a). Particles with smaller size induced higher aggregations of neutrophils and apoptosis of macrophages in the abdomen of the larvae, corresponding to greater hepatic inflammation in the larvae. NPs increased the expression of fabp10a in the larval livers in a dose- and size-dependent manner. PS particles of 50 nm at a concentration of 0.1 mg·L-1 increased the expression of fabp10a in the larval liver by 21.90% (P < 0.05). The plausible mechanisms of these effects depend on their distribution and the generation of reactive oxygen species in the larvae. Metabonomic analysis revealed that the metabolic pathways of catabolic processes, amino acids, and purines were highly promoted by NPs, compared to micron PS particles. NPs also activate steroid hormone biosynthesis in zebrafish larvae, which may lead to the occurrence of immune-related diseases. For the first time, the liver was identified as the target organ for the immunotoxicity effects of NPs in the larval stage of fish.
Collapse
Affiliation(s)
- Haodong Cheng
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology / School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Zhenghua Duan
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology / School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China; MOE Key Laboratory on Pollution Processes and Environmental Criteria / College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| | - Yinghong Wu
- Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Yudi Wang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology / School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Haihong Zhang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology / School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Yansong Shi
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology / School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Huajing Zhang
- MOE Key Laboratory on Pollution Processes and Environmental Criteria / College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Yanjie Wei
- Key Laboratory of Environmental Protection in Water Transport Engineering Ministry of Communications, Tianjin Research Institute of Water Transport Engineering, Tianjin 300456, China
| | - Hongwen Sun
- MOE Key Laboratory on Pollution Processes and Environmental Criteria / College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
23
|
Qin L, Duan Z, Cheng H, Wang Y, Zhang H, Zhu Z, Wang L. Size-dependent impact of polystyrene microplastics on the toxicity of cadmium through altering neutrophil expression and metabolic regulation in zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 291:118169. [PMID: 34536643 DOI: 10.1016/j.envpol.2021.118169] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 06/13/2023]
Abstract
Insufficient evidence exists regarding the visible physiological toxic endpoints of MPs exposures on zebrafish larvae due to their small sizes. Herein, the impacts of micro-polystyrene particles (μ-PS) and 100 nm polystyrene particles (n-PS) on the toxicity of cadmium (Cd) through altering neutrophil expressions were identified and quantified in the transgenic zebrafish (Danio rerio) larvae Tg(lyz:DsRed2), and the effects were size-dependent. When exposed together with μ-PS, the amount of neutrophils in Cd treated zebrafish larvae decreased by 25.56% through reducing Cd content in the larvae. By contrast, although n-PS exposure caused lower Cd content in the larvae, the expression of neutrophils under their combined exposure remained high. The mechanism of immune toxicity was analyzed based on the results of metabonomics. n-PS induced high oxidative stress in the larvae, which promoted taurine metabolism and unsaturated fatty biosynthesis in n-PS + Cd treatment. This observation was accordance with the significant inhibition of the activities of superoxide dismutase and catalase enzymes detected in their combined treatment. Moreover, n-PS promoted the metabolic pathways of catabolic processes, amino acid metabolism, purine metabolism, and steroid hormone biosynthesis in Cd treated zebrafish larvae. Nanoplasctis widely coexist with other pollutants in the environment at relatively low concentrations. We conclude that more bio-markers of immune impact should be explored to identify their toxicological mechanisms and mitigate the effects on the environment.
Collapse
Affiliation(s)
- Li Qin
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Zhenghua Duan
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, Tianjin University of Technology, Tianjin, 300384, China.
| | - Haodong Cheng
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, Tianjin University of Technology, Tianjin, 300384, China
| | - Yudi Wang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, Tianjin University of Technology, Tianjin, 300384, China
| | - Haihong Zhang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, Tianjin University of Technology, Tianjin, 300384, China
| | - Zhe Zhu
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, Tianjin University of Technology, Tianjin, 300384, China
| | - Lei Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Nankai University, Tianjin, 300071, China
| |
Collapse
|
24
|
Lubin A, Otterstrom J, Hoade Y, Bjedov I, Stead E, Whelan M, Gestri G, Paran Y, Payne E. A versatile, automated and high-throughput drug screening platform for zebrafish embryos. Biol Open 2021; 10:bio058513. [PMID: 34472582 PMCID: PMC8430230 DOI: 10.1242/bio.058513] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/28/2021] [Indexed: 12/29/2022] Open
Abstract
Zebrafish provide a unique opportunity for drug screening in living animals, with the fast-developing, transparent embryos allowing for relatively high-throughput, microscopy-based screens. However, the limited availability of rapid, flexible imaging and analysis platforms has limited the use of zebrafish in drug screens. We have developed an easy-to-use, customisable automated screening procedure suitable for high-throughput phenotype-based screens of live zebrafish. We utilised the WiScan® Hermes High Content Imaging System to rapidly acquire brightfield and fluorescent images of embryos, and the WiSoft® Athena Zebrafish Application for analysis, which harnesses an Artificial Intelligence-driven algorithm to automatically detect fish in brightfield images, identify anatomical structures, partition the animal into regions and exclusively select the desired side-oriented fish. Our initial validation combined structural analysis with fluorescence images to enumerate GFP-tagged haematopoietic stem and progenitor cells in the tails of embryos, which correlated with manual counts. We further validated this system to assess the effects of genetic mutations and X-ray irradiation in high content using a wide range of assays. Further, we performed simultaneous analysis of multiple cell types using dual fluorophores in high throughput. In summary, we demonstrate a broadly applicable and rapidly customisable platform for high-content screening in zebrafish. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Alexandra Lubin
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| | | | - Yvette Hoade
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Ivana Bjedov
- Research Department of Cancer Biology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Eleanor Stead
- Research Department of Cancer Biology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Matthew Whelan
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Gaia Gestri
- Department of Cell and Developmental Biology, University College London, London WC1E 6AR, UK
| | - Yael Paran
- IDEA Bio-Medical Ltd., Rehovot 76705, Israel
| | - Elspeth Payne
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| |
Collapse
|
25
|
Peña OA, Lubin A, Rowell J, Hoade Y, Khokhar N, Lemmik H, Mahony C, Dace P, Umamahesan C, Payne EM. Differential Requirement of Gata2a and Gata2b for Primitive and Definitive Myeloid Development in Zebrafish. Front Cell Dev Biol 2021; 9:708113. [PMID: 34589480 PMCID: PMC8475954 DOI: 10.3389/fcell.2021.708113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/10/2021] [Indexed: 11/14/2022] Open
Abstract
Germline loss or mutation of one copy of the transcription factor GATA2 in humans leads to a range of clinical phenotypes affecting hematopoietic, lymphatic and vascular systems. GATA2 heterozygous mice show only a limited repertoire of the features observed in humans. Zebrafish have two copies of the Gata2 gene as a result of an additional round of ancestral whole genome duplication. These genes, Gata2a and Gata2b, show distinct but overlapping expression patterns, and between them, highlight a significantly broader range of the phenotypes observed in GATA2 deficient syndromes, than each one alone. In this manuscript, we use mutants for Gata2a and Gata2b to interrogate the effects on hematopoiesis of these two ohnologs, alone and in combination, during development in order to further define the role of GATA2 in developmental hematopoiesis. We define unique roles for each ohnolog at different stages of developmental myelopoiesis and for the emergence of hematopoietic stem and progenitor cells. These effects are not additive in the haploinsufficient state suggesting a redundancy between these two genes in hematopoietic stem and progenitor cells. Rescue studies additionally support that Gata2b can compensate for the effects of Gata2a loss. Finally we show that adults with loss of combined heterozygosity show defects in the myeloid compartment consistent with GATA2 loss in humans. These results build on existing knowledge from other models of GATA2 deficiency and refine our understanding of the early developmental effects of GATA2. In addition, these studies shed light on the complexity and potential structure-function relationships as well as sub-functionalization of Gata2 genes in the zebrafish model.
Collapse
Affiliation(s)
- Oscar A. Peña
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Alexandra Lubin
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Jasmine Rowell
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Yvette Hoade
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Noreen Khokhar
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Hanna Lemmik
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Christopher Mahony
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Phoebe Dace
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Chianna Umamahesan
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Elspeth M. Payne
- Research Department of Haematology, Cancer Institute, University College London, London, United Kingdom
- National Institute for Health Research (NIHR)/UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
26
|
Isles HM, Loynes CA, Alasmari S, Kon FC, Henry KM, Kadochnikova A, Hales J, Muir CF, Keightley MC, Kadirkamanathan V, Hamilton N, Lieschke GJ, Renshaw SA, Elks PM. Pioneer neutrophils release chromatin within in vivo swarms. eLife 2021; 10:68755. [PMID: 34292151 PMCID: PMC8298094 DOI: 10.7554/elife.68755] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022] Open
Abstract
Neutrophils are rapidly recruited to inflammatory sites where their coordinated migration forms clusters, a process termed neutrophil swarming. The factors that modulate early stages of neutrophil swarming are not fully understood, requiring the development of new in vivo models. Using transgenic zebrafish larvae to study endogenous neutrophil migration in a tissue damage model, we demonstrate that neutrophil swarming is a conserved process in zebrafish immunity, sharing essential features with mammalian systems. We show that neutrophil swarms initially develop around an individual pioneer neutrophil. We observed the violent release of extracellular cytoplasmic and nuclear fragments by the pioneer and early swarming neutrophils. By combining in vitro and in vivo approaches to study essential components of neutrophil extracellular traps (NETs), we provide in-depth characterisation and high-resolution imaging of the composition and morphology of these release events. Using a photoconversion approach to track neutrophils within developing swarms, we identify that the fate of swarm-initiating pioneer neutrophils involves extracellular chromatin release and that the key NET components gasdermin, neutrophil elastase, and myeloperoxidase are required for the swarming process. Together our findings demonstrate that release of cellular components by pioneer neutrophils is an initial step in neutrophil swarming at sites of tissue injury.
Collapse
Affiliation(s)
- Hannah M Isles
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Catherine A Loynes
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Sultan Alasmari
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Fu Chuen Kon
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Katherine M Henry
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Anastasia Kadochnikova
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Jack Hales
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Clare F Muir
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | | | - Visakan Kadirkamanathan
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Noémie Hamilton
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Stephen A Renshaw
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Philip M Elks
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| |
Collapse
|
27
|
Silva NV, Carregosa D, Gonçalves C, Vieira OV, Nunes Dos Santos C, Jacinto A, Crespo CL. A Dietary Cholesterol-Based Intestinal Inflammation Assay for Improving Drug-Discovery on Inflammatory Bowel Diseases. Front Cell Dev Biol 2021; 9:674749. [PMID: 34150769 PMCID: PMC8209420 DOI: 10.3389/fcell.2021.674749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBD) with chronic infiltration of immune cells in the gastrointestinal tract are common and largely incurable. The therapeutic targeting of IBD has been hampered by the complex causality of the disease, with environmental insults like cholesterol-enriched Western diets playing a critical role. To address this drug development challenge, we report an easy-to-handle dietary cholesterol-based in vivo assay that allows the screening of immune-modulatory therapeutics in transgenic zebrafish models. An improvement in the feeding strategy with high cholesterol diet (HCD) selectively induces a robust and consistent infiltration of myeloid cells in larvae intestines that is highly suitable for compound discovery efforts. Using transgenics with fluorescent reporter expression in neutrophils, we take advantage of the unique zebrafish larvae clarity to monitor an acute inflammatory response in a whole organism context with a fully functional innate immune system. The use of semi-automated image acquisition and processing combined with quantitative image analysis allows categorizing anti- or pro-inflammatory compounds based on a leukocytic inflammation index. Our HCD gut inflammation (HCD-GI) assay is simple, cost- and time-effective as well as highly physiological which makes it unique when compared to chemical-based zebrafish models of IBD. Besides, diet is a highly controlled, selective and targeted trigger of intestinal inflammation that avoids extra-intestinal outcomes and reduces the chances of chemical-induced toxicity during screenings. We show the validity of this assay for a screening platform by testing two dietary phenolic acids, namely gallic acid (GA; 3,4,5-trihydroxybenzoic acid) and ferulic acid (FA; 4-hydroxy-3-methoxycinnamic acid), with well described anti-inflammatory actions in animal models of IBD. Analysis of common IBD therapeutics (Prednisolone and Mesalamine) proved the fidelity of our IBD-like intestinal inflammation model. In conclusion, the HCD-GI assay can facilitate and accelerate drug discovery efforts on IBD, by identification of novel lead molecules with immune modulatory action on intestinal neutrophilic inflammation. This will serve as a jumping-off point for more profound analyses of drug mechanisms and pathways involved in early IBD immune responses.
Collapse
Affiliation(s)
- Nuno-Valério Silva
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Diogo Carregosa
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Catarina Gonçalves
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal.,Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - António Jacinto
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Carolina Lage Crespo
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
28
|
Gibson JF, Prajsnar TK, Hill CJ, Tooke AK, Serba JJ, Tonge RD, Foster SJ, Grierson AJ, Ingham PW, Renshaw SA, Johnston SA. Neutrophils use selective autophagy receptor Sqstm1/p62 to target Staphylococcus aureus for degradation in vivo in zebrafish. Autophagy 2021; 17:1448-1457. [PMID: 32559122 PMCID: PMC8204994 DOI: 10.1080/15548627.2020.1765521] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 04/24/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022] Open
Abstract
Macroautophagy/autophagy functions to degrade cellular components and intracellular pathogens. Autophagy receptors, including SQSTM1/p62, target intracellular pathogens. Staphylococcus aureus is a significant pathogen of humans, especially in immunocompromise. S. aureus may use neutrophils as a proliferative niche, but their intracellular fate following phagocytosis has not been analyzed in vivo. In vitro, SQSTM1 can colocalize with intracellular Staphylococcus aureus, but whether SQSTM1 is beneficial or detrimental in host defense against S. aureus in vivo is unknown. Here we determine the fate and location of S. aureus within neutrophils throughout zebrafish infection. We show Lc3 and Sqstm1 recruitment to phagocytosed S. aureus is altered depending on the bacterial location within the neutrophil and that Lc3 marking of bacterial phagosomes within neutrophils may precede bacterial degradation. Finally, we show Sqstm1 is important for controlling cytosolic bacteria, demonstrating for the first time a key role of Sqstm1 in autophagic control of S. aureus in neutrophils.Abbreviations: AR: autophagy receptor; CFU: colony-forming unit; CHT: caudal hematopoietic tissue; GFP: green fluorescent protein; hpf: hours post-fertilization; hpi: hours post-infection; LWT: london wild-type: lyz: lysozyme; Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; RFP: red fluorescent protein; Sqstm1/p62: sequestosome 1; Tg: transgenic; TSA: tyramide signal amplification; UBD: ubiquitin binding domain.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-star), Singapore
- Florey Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Tomasz K. Prajsnar
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Institute Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Christopher J. Hill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Amy K. Tooke
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Justyna J. Serba
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
| | - Rebecca D. Tonge
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Simon J Foster
- Florey Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Andrew J. Grierson
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Philip W. Ingham
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-star), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| | - Simon A. Johnston
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
29
|
Chen W, Xie L, Yu F, Li Y, Chen C, Xie W, Huang T, Zhang Y, Zhang S, Li P. Zebrafish as a Model for In-Depth Mechanistic Study for Stroke. Transl Stroke Res 2021; 12:695-710. [PMID: 34050491 DOI: 10.1007/s12975-021-00907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022]
Abstract
Stroke is one of the world's leading causes of death and disability, posing enormous burden to the society. However, the pathogenesis and mechanisms that underlie brain injury and brain repair remain largely unknown. There's an unmet need of in-depth mechanistic research in this field. Zebrafish (Danio rerio) is a powerful tool in brain science research mainly due to its small size and transparent body, high genome synteny with human, and similar nervous system structures. It can be used to establish both hemorrhagic and ischemic stroke models easily and effectively through different ways. After the establishment of stroke model, research methods including behavioral test, in vivo imaging, and drug screening are available to explore mechanisms that underlie the brain injury and brain repair after stroke. This review focuses on the advantages and the feasibility of zebrafish stroke model, and will also introduce the key methods available for stroke studies in zebrafish, which may drive future mechanistic studies in the pursuit of discovering novel therapeutic targets for stroke patients.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Fang Yu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Wanqing Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Song Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
30
|
Shameena SS, Kumar S, Kumar K, Raman RP. Role of temperature and co-infection in mediating the immune response of goldfish. Microb Pathog 2021; 156:104896. [PMID: 33965506 DOI: 10.1016/j.micpath.2021.104896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
Aquatic Pathogens are expected to encounter tremendous levels of variation in their environment - both abiotic and biotic. Here we examined the change in innate immune parameters and mortality pattern of Carassius auratus during the interaction of co-infection due to an ectoparasite, Argulus and bacteria Aeromonas hydrophila, along with a temperature gradient. Experimental fish were assigned randomly to six treatment groups (T1-T6). Fish of groups T1, T3 and T5 are assigned for healthy fishes kept at 23, 28 and 33°c temperature and served as control. T2, T4 and T6 groups are the co-infected groups kept at temperature gradient. For the haematological and enzyme parameter analysis, sampling was done at 24 h, 72 h and 168 h post challenge from 4 fish in all experimental groups. A temperature dependent increase in intensity of Argulus was observed in the experimental group. Both in control group and co-infected group a temperature dependent mortality pattern was observed, showing an increased mortality of 60% in T6 and 20% in T5 group. A significant decrease of RBC, Hb, and PCV values was observed in co-infected group when compared with control fish in each of the experimental group. Also a temperature dependent increase in WBC, neutrophil and monocyte value was observed in control fish. Whereas, a significant reduction in WBC, neutrophil and monocyte was observed in co-infected fish exposed to 33 °C during the progression of infection. Furthermore, T4 group showed a significantly higher Nitroblue tetrazolium test, Myelo peroxidase and lyzozyme activity compared to other co-infection group. A significantly increased activity of Superoxide dismutase, Glutathione peroxidise and catalase activity was recorded in control fish exposed to 33 °C (T5) whereas, there was no significant difference observed in the activity of catalase and Glutathione peroxidise in the other control fish (T1 and T3 group). This result implies that increase in temperature not only accelerates the intensity of co-infection but also imbalance the health status of the fish by hampering the immunological and physiological parameters towards more detrimental side.
Collapse
Affiliation(s)
- S S Shameena
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, Maharashtra, India
| | - Saurav Kumar
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, Maharashtra, India
| | - Kundan Kumar
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, Maharashtra, India
| | - R P Raman
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, Maharashtra, India.
| |
Collapse
|
31
|
Zhao Y, Zhou L, Li H, Sun T, Wen X, Li X, Meng Y, Li Y, Liu M, Liu S, Kim SJ, Xiao J, Li L, Zhang S, Li W, Cohen P, Hoffman AR, Hu JF, Cui J. Nuclear-Encoded lncRNA MALAT1 Epigenetically Controls Metabolic Reprogramming in HCC Cells through the Mitophagy Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:264-276. [PMID: 33425485 PMCID: PMC7773746 DOI: 10.1016/j.omtn.2020.09.040] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction is a metabolic hallmark of cancer cells. In search of molecular factors involved in this dysregulation in hepatocellular carcinoma (HCC), we found that the nuclear-encoded long noncoding RNA (lncRNA) MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) was aberrantly enriched in the mitochondria of hepatoma cells. Using RNA reverse transcription-associated trap sequencing (RAT-seq), we showed that MALAT1 interacted with multiple loci on mitochondrial DNA (mtDNA), including D-loop, COX2, ND3, and CYTB genes. MALAT1 knockdown induced alterations in the CpG methylation of mtDNA and in mitochondrial transcriptomes. This was associated with multiple abnormalities in mitochondrial function, including altered mitochondrial structure, low oxidative phosphorylation (OXPHOS), decreased ATP production, reduced mitophagy, decreased mtDNA copy number, and activation of mitochondrial apoptosis. These alterations in mitochondrial metabolism were associated with changes in tumor phenotype and in pathways involved in cell mitophagy, mitochondrial apoptosis, and epigenetic regulation. We further showed that the RNA-shuttling protein HuR and the mitochondria transmembrane protein MTCH2 mediated the transport of MALAT1 in this nuclear-mitochondrial crosstalk. This study provides the first evidence that the nuclear genome-encoded lncRNA MALAT1 functions as a critical epigenetic player in the regulation of mitochondrial metabolism of hepatoma cells, laying the foundation for further clarifying the roles of lncRNAs in tumor metabolic reprogramming.
Collapse
Affiliation(s)
- Yijing Zhao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Lei Zhou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Hui Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Tingge Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xue Wen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xueli Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Yan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Mengmeng Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Shanshan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Su-Jeong Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jialin Xiao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Lingyu Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Songling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew R. Hoffman
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Ji-Fan Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Medicine, PAVIR, Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Jiuwei Cui
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
32
|
Buchan KD, van Gent M, Prajsnar TK, Ogryzko NV, de Jong NWM, Kolata J, Foster SJ, van Strijp JAG, Renshaw SA. Human-specific staphylococcal virulence factors enhance pathogenicity in a humanised zebrafish C5a receptor model. J Cell Sci 2021; 134:jcs.252205. [PMID: 33589501 DOI: 10.1242/jcs.252205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/12/2021] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus infects ∼30% of the human population and causes a spectrum of pathologies ranging from mild skin infections to life-threatening invasive diseases. The strict host specificity of its virulence factors has severely limited the accuracy of in vivo models for the development of vaccines and therapeutics. To resolve this, we generated a humanised zebrafish model and determined that neutrophil-specific expression of the human C5a receptor conferred susceptibility to the S. aureus toxins PVL and HlgCB, leading to reduced neutrophil numbers at the site of infection and increased infection-associated mortality. These results show that humanised zebrafish provide a valuable platform to study the contribution of human-specific S. aureus virulence factors to infection in vivo that could facilitate the development of novel therapeutic approaches and essential vaccines.
Collapse
Affiliation(s)
- Kyle D Buchan
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.,The Florey Institute for Host-Pathogen Interactions, Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Michiel van Gent
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CX, The Netherlands
| | - Tomasz K Prajsnar
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Nikolay V Ogryzko
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Nienke W M de Jong
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CX, The Netherlands
| | - Julia Kolata
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CX, The Netherlands
| | - Simon J Foster
- The Florey Institute for Host-Pathogen Interactions, Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CX, The Netherlands
| | - Stephen A Renshaw
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
33
|
Paredes LC, Luz RBDS, Tozzi ON, de Carvalho LÂSJ, Calado SLDM, Padovani BN, Fénero CIM, do Amaral MA, de Assis HCDS, Câmara NOS, Braga TT. Distinct macrophage phenotypes and redox environment during the fin fold regenerative process in zebrafish. Scand J Immunol 2021; 94:e13026. [PMID: 33565093 DOI: 10.1111/sji.13026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 12/15/2022]
Abstract
In contrast to mammals, zebrafish (Danio rerio) has the ability to regenerate injured sites such as different tissues present in the fin. It is known that cells of the innate immune system play essential roles in regeneration; however, some aspects of the molecular mechanisms by which these cells orchestrate regeneration remain unknown. This study aimed to evaluate the infiltration dynamics of neutrophils and macrophages in the regenerative process of fin fold in regard to the influence of the redox environment and oxidative pathways. Fin fold amputation was performed on transgenic larvae for macrophage-expressed gene 1 (mpeg1), lysozyme (lyz), myeloperoxidase (mpo) and tumour necrosis factor alpha (TNFα) at 3 days post-fertilization, followed by confocal microscopy imaging and measurement of the activities of oxidant and antioxidant enzymes. We observed initially an increase in the number of neutrophils (lyz:DsRed+/mpx:GFP+) and then macrophages (mpeg1+) in the injury site followed by a decrease in neutrophils at 7 days post-amputation (dpa). Moreover, macrophages switch from a pro-inflammatory to an anti-inflammatory profile throughout the process, while the activity of superoxide dismutase (SOD) increased at 1 dpa and catalase (CAT) at 5 dpa. Higher levels of lipid peroxidation were also detected during regeneration. Despite oxidative stress, there is, therefore, an antioxidant response throughout the regeneration of the caudal fin. The present work can contribute to future studies on the development of cell therapies, achieving greater effectiveness in the treatment of diseases related to the formation of fibrotic tissue.
Collapse
Affiliation(s)
| | | | | | | | | | - Bárbara Nunes Padovani
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil
| | | | - Mariana Abrantes do Amaral
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil
| | | | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo, São Paulo, Brazil.,Nephrology Division, Federal University of São Paulo, São Paulo, Brazil
| | - Tarcio Teodoro Braga
- Department of Pathology, Federal University of Parana, Curitiba, Brazil.,Graduate Program in Biosciences and Biotechnology, Instituto Carlos Chagas, Fiocruz-Parana, Curitiba, Brazil
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Myeloid cells contribute to immune response to infection and tissue regeneration after injury as well as to the developmental induction of the hematopoietic system overall. Here we review recent uses of zebrafish to advance the study of myeloid biology in development and disease. RECENT FINDINGS Recent studies have made use of advanced imaging and genetic strategies and have highlighted key concepts in myeloid cell behavior. These include immune-cell cross-talk and subpopulation response in infection and regeneration, and tightly regulated inflammatory and tissue remodeling behaviors in development. SUMMARY These new findings will shape our understanding of the developmental origins of immune populations as well as their specific cellular behaviors at all stages of infection, regeneration, and myeloid neoplasms.
Collapse
Affiliation(s)
- Samuel J. Wattrus
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The availability of organs for transplant fails to meet the demand and this shortage is growing worse every year. As the cost of not getting a suitable donor organ can mean death for patients, new tools and approaches that allows us to make advances in transplantation faster and provide a different vantage point are required. To address this need, we introduce the concept of using the zebrafish (Danio rerio) as a new model system in organ transplantation. The zebrafish community offers decades of research experience in disease modeling and a rich toolbox of approaches for interrogating complex pathological states. We provide examples of how already existing zebrafish assays/tools from cancer, regenerative medicine, immunology, and others, could be leveraged to fuel new discoveries in pursuit of solving the organ shortage. RECENT FINDINGS Important innovations have enabled several types of transplants to be successfully performed in zebrafish, including stem cells, tumors, parenchymal cells, and even a partial heart transplant. These innovations have been performed against a backdrop of an expansive and impressive list of tools designed to uncover the biology of complex systems that include a wide array of fluorescent transgenic fish that label specific cell types and mutant lines that are transparent, immune-deficient. Allogeneic transplants can also be accomplished using immune suppressed and syngeneic fish. Each of these innovations within the zebrafish community would provide several helpful tools that could be applied to transplant research. SUMMARY We highlight some examples of existing tools and assays developed in the zebrafish community that could be leveraged to overcome barriers in organ transplantation, including ischemia-reperfusion, short preservation durations, regeneration of marginal grafts, and acute and chronic rejection.
Collapse
|
36
|
López Nadal A, Ikeda-Ohtsubo W, Sipkema D, Peggs D, McGurk C, Forlenza M, Wiegertjes GF, Brugman S. Feed, Microbiota, and Gut Immunity: Using the Zebrafish Model to Understand Fish Health. Front Immunol 2020; 11:114. [PMID: 32117265 PMCID: PMC7014991 DOI: 10.3389/fimmu.2020.00114] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Aquafeed companies aim to provide solutions to the various challenges related to nutrition and health in aquaculture. Solutions to promote feed efficiency and growth, as well as improving the fish health or protect the fish gut from inflammation may include dietary additives such as prebiotics and probiotics. The general assumption is that feed additives can alter the fish microbiota which, in turn, interacts with the host immune system. However, the exact mechanisms by which feed influences host-microbe-immune interactions in fish still remain largely unexplored. Zebrafish rapidly have become a well-recognized animal model to study host-microbe-immune interactions because of the diverse set of research tools available for these small cyprinids. Genome editing technologies can create specific gene-deficient zebrafish that may contribute to our understanding of immune functions. Zebrafish larvae are optically transparent, which allows for in vivo imaging of specific (immune) cell populations in whole transgenic organisms. Germ-free individuals can be reared to study host-microbe interactions. Altogether, these unique zebrafish features may help shed light on the mechanisms by which feed influences host-microbe-immune interactions and ultimately fish health. In this review, we first describe the anatomy and function of the zebrafish gut: the main surface where feed influences host-microbe-immune interactions. Then, we further describe what is currently known about the molecular pathways that underlie this interaction in the zebrafish gut. Finally, we summarize and critically review most of the recent research on prebiotics and probiotics in relation to alterations of zebrafish microbiota and immune responses. We discuss the advantages and disadvantages of the zebrafish as an animal model for other fish species to study feed effects on host-microbe-immune interactions.
Collapse
Affiliation(s)
- Adrià López Nadal
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, Netherlands.,Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, Netherlands
| | - Wakako Ikeda-Ohtsubo
- Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Detmer Sipkema
- Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - David Peggs
- Skretting Aquaculture Research Centre, Stavanger, Norway
| | - Charles McGurk
- Skretting Aquaculture Research Centre, Stavanger, Norway
| | - Maria Forlenza
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, Netherlands
| | - Geert F Wiegertjes
- Aquaculture and Fisheries Group, Wageningen University and Research, Wageningen, Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
37
|
Flores EM, Nguyen AT, Odem MA, Eisenhoffer GT, Krachler AM. The zebrafish as a model for gastrointestinal tract-microbe interactions. Cell Microbiol 2020; 22:e13152. [PMID: 31872937 DOI: 10.1111/cmi.13152] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
The zebrafish (Danio rerio) has become a widely used vertebrate model for bacterial, fungal, viral, and protozoan infections. Due to its genetic tractability, large clutch sizes, ease of manipulation, and optical transparency during early life stages, it is a particularly useful model to address questions about the cellular microbiology of host-microbe interactions. Although its use as a model for systemic infections, as well as infections localised to the hindbrain and swimbladder having been thoroughly reviewed, studies focusing on host-microbe interactions in the zebrafish gastrointestinal tract have been neglected. Here, we summarise recent findings regarding the developmental and immune biology of the gastrointestinal tract, drawing parallels to mammalian systems. We discuss the use of adult and larval zebrafish as models for gastrointestinal infections, and more generally, for studies of host-microbe interactions in the gut.
Collapse
Affiliation(s)
- Erika M Flores
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Anh T Nguyen
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - George T Eisenhoffer
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.,M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
38
|
Jiang A, Zhang Y, Zhang X, Wu D, Liu Z, Li S, Liu X, Han Z, Wang C, Wang J, Wei Z, Guo C, Yang Z. Morin alleviates LPS-induced mastitis by inhibiting the PI3K/AKT, MAPK, NF-κB and NLRP3 signaling pathway and protecting the integrity of blood-milk barrier. Int Immunopharmacol 2019; 78:105972. [PMID: 31711938 DOI: 10.1016/j.intimp.2019.105972] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Mastitis is a common veterinary clinical disease that restricts the development of dairy farming around the world. Morin, extracted from Mulberry Tree and other herbs, has been reported to possess the function of anti-bacteria, anti-oxidant, and anti-inflammatory. However, whether morin could protect lipopolysaccharide (LPS)-induced mouse mastitis in vivo has not well known. This study firstly aims to evaluate the effects of morin on LPS-induced mouse mastitis in vivo, and then try to illustrate the mechanism involved in the process. Before injected with LPS, mice were intraperitoneally pre-injected with different concentrations of morin, and mice of the control and LPS group were injected with the same amount of saline. Pathologic changes of mammary gland were determined by histopathological examination. Myeloperoxidase (MPO) activities of mammary gland were determined by the MPO kits. The mRNA expressions of inflammatory cytokines including TNF-α, IL-1β and IL-6, and those of chemokine factors CCL2 and CXCL2, and those of tight junctions occludin claudin-3 were examined by qRT-PCR analysis. The activities of IκB, p65, ERK, P38, AKT, PI3K, NLPR3, claudin-1, claudin-3 and occludin were determined by western blotting. The results showed that morin alleviated LPS-induced edema, destructed structures and infiltrated inflammatory cells of mammary gland. Morin administration significantly decreased LPS-induced TNF-α, IL-1β, IL-6, CCL2 and CXCL2 mRNA expressions. Furthermore, western blot analysis also showed that morin significantly reduced LPS-induced phosphorylation of p65, IκB, p38 and ERK, and enhanced LPS-induced phosphorylation of AKT and PI3K. It was also found that LPS-decreased claudin-3 and occludin expressions were also inhibited by morin treatment. In summary, above results suggest that morin indeed protect LPS-induced mouse mastitis in vivo, and the mechanism was through inhibiting the PI3K/AKT, MAPK, NF-κB and NLRP3 signaling pathways and protecting the integrity of blood-milk barrier by regulating the tight junction proteins expressions.
Collapse
Affiliation(s)
- Aimin Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Yong Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Xu Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Di Wu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Ziyi Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Shuangqiu Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Xiao Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Zhen Han
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Chaoqun Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Jingjing Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Zhengkai Wei
- College of Life Sciences and Engineering, Foshan University Foshan 528225, Guangdong Province, PR China
| | - Changming Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China
| | - Zhengtao Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, PR China; College of Life Sciences and Engineering, Foshan University Foshan 528225, Guangdong Province, PR China.
| |
Collapse
|
39
|
Murdoch CC, Rawls JF. Commensal Microbiota Regulate Vertebrate Innate Immunity-Insights From the Zebrafish. Front Immunol 2019; 10:2100. [PMID: 31555292 PMCID: PMC6742977 DOI: 10.3389/fimmu.2019.02100] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
Microbial communities populate the mucosal surfaces of all animals. Metazoans have co-evolved with these microorganisms, forming symbioses that affect the molecular and cellular underpinnings of animal physiology. These microorganisms, collectively referred to as the microbiota, are found on many distinct body sites (including the skin, nasal cavity, and urogenital tract), however the most densely colonized host tissue is the intestinal tract. Although spatially confined within the intestinal lumen, the microbiota and associated products shape the development and function of the host immune system. Studies comparing gnotobiotic animals devoid of any microbes (germ free) with counterparts colonized with selected microbial communities have demonstrated that commensal microorganisms are required for the proper development and function of the immune system at homeostasis and following infectious challenge or injury. Animal model systems have been essential for defining microbiota-dependent shifts in innate immune cell function and intestinal physiology during infection and disease. In particular, the zebrafish has emerged as a powerful vertebrate model organism with unparalleled capacity for in vivo imaging, a full complement of genetic approaches, and facile methods to experimentally manipulate microbial communities. Here we review key insights afforded by the zebrafish into the impact of microbiota on innate immunity, including evidence that the perception of and response to the microbiota is evolutionarily conserved. We also highlight opportunities to strengthen the zebrafish model system, and to gain new insights into microbiota-innate immune interactions that would be difficult to achieve in mammalian models.
Collapse
Affiliation(s)
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|