1
|
McGrew MJ, Holmes T, Davey MG. A scientific case for revisiting the embryonic chicken model in biomedical research. Dev Biol 2025; 522:220-226. [PMID: 40015500 DOI: 10.1016/j.ydbio.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
The availability of fertilised chicken eggs and the accessibility and rapid development of the avian embryo, have been utilised in biomedical scientific research to make fundamental discoveries including of developmental processes that are common to all vertebrates, advances in teratology, the understanding of tumour growth and metastasis, angiogenesis, cancer drug assessment and vaccine development as well as advances in understanding avian specific biology. However, recent innovations in chicken transgenesis, genome engineering and surrogate host technology in chickens have only been utilised in a few of these fields of research, specifically some areas of developmental biology, avian sex determination and immunology. To understand why other biomedical fields have not adopted modern chicken transgenic tools, we investigated the non-technical summaries of projects granted in the UK under the Animals (Scientific Procedures) Act 1986 between 2017 and 2023 to assess when and how chicken embryos are used in research, and if they were considered as a Replacement model for other species.
Collapse
Affiliation(s)
- Mike J McGrew
- Division of Translational Bioscience, The Roslin Institute and R(D)SVS, University of Edinburgh, EH25 9RG, UK; Roslin Institute Chicken Embryology (RICE) Group, UK
| | - Tana Holmes
- Division of Functional Genetics, The Roslin Institute and R(D)SVS, University of Edinburgh, EH25 9RG, UK
| | - Megan G Davey
- Division of Functional Genetics, The Roslin Institute and R(D)SVS, University of Edinburgh, EH25 9RG, UK; Roslin Institute Chicken Embryology (RICE) Group, UK.
| |
Collapse
|
2
|
Guan D, Bai Z, Zhu X, Zhong C, Hou Y, Zhu D, Li H, Lan F, Diao S, Yao Y, Zhao B, Li X, Pan Z, Gao Y, Wang Y, Zou D, Wang R, Xu T, Sun C, Yin H, Teng J, Xu Z, Lin Q, Shi S, Shao D, Degalez F, Lagarrigue S, Wang Y, Wang M, Peng M, Rocha D, Charles M, Smith J, Watson K, Buitenhuis AJ, Sahana G, Lund MS, Warren W, Frantz L, Larson G, Lamont SJ, Si W, Zhao X, Li B, Zhang H, Luo C, Shu D, Qu H, Luo W, Li Z, Nie Q, Zhang X, Xiang R, Liu S, Zhang Z, Zhang Z, Liu GE, Cheng H, Yang N, Hu X, Zhou H, Fang L. Genetic regulation of gene expression across multiple tissues in chickens. Nat Genet 2025:10.1038/s41588-025-02155-9. [PMID: 40200121 DOI: 10.1038/s41588-025-02155-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
The chicken is a valuable model for understanding fundamental biology and vertebrate evolution and is a major global source of nutrient-dense and lean protein. Despite being the first non-mammalian amniote to have its genome sequenced, a systematic characterization of functional variation on the chicken genome remains lacking. Here, we integrated bulk RNA sequencing (RNA-seq) data from 7,015 samples, single-cell RNA-seq data from 127,598 cells and 2,869 whole-genome sequences to present a pilot atlas of regulatory variants across 28 chicken tissues. This atlas reveals millions of regulatory effects on primary expression (protein-coding genes, long non-coding RNA and exons) and post-transcriptional modifications (alternative splicing and 3'-untranslated region alternative polyadenylation). We highlighted distinct molecular mechanisms underlying these regulatory variants, their context-dependent behavior and their utility in interpreting genome-wide associations for 39 chicken complex traits. Finally, our comparative analyses of gene regulation between chickens and mammals demonstrate how this resource can facilitate cross-species gene mapping of complex traits.
Collapse
Affiliation(s)
- Dailu Guan
- Department of Animal Science, University of California-Davis, Davis, CA, USA
| | - Zhonghao Bai
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Xiaoning Zhu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Conghao Zhong
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yali Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Di Zhu
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Houcheng Li
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuqi Diao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yuelin Yao
- MRC Human Genetics Unit at the Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- School of Informatics, The University of Edinburgh, Edinburgh, UK
| | - Bingru Zhao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhangyuan Pan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yahui Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Yuzhe Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dong Zou
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Ruizhen Wang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianyi Xu
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hongwei Yin
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Jinyan Teng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhiting Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qing Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shourong Shi
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Dan Shao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | | | | | - Ying Wang
- Department of Animal Science, University of California-Davis, Davis, CA, USA
| | - Mingshan Wang
- State Key Laboratory of Genetic Evolution & Animal Models and Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Minsheng Peng
- State Key Laboratory of Genetic Evolution & Animal Models and Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Dominique Rocha
- INRAE, GABI, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mathieu Charles
- INRAE, GABI, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jacqueline Smith
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Kellie Watson
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | | | - Goutam Sahana
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Wesley Warren
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, MO, USA
| | - Laurent Frantz
- Palaeogenomics Group, Department of Veterinary Sciences, Ludwig Maximilian University, Munich, Germany
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Greger Larson
- The Palaeogenomics & Bio-Archaeology Research Network, School of Archaeology, University of Oxford, Oxford, UK
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Wei Si
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Animal Science, McGill University, Montreal, Quebec, Canada
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, Quebec, Canada
| | - Bingjie Li
- Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, UK
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chenglong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Dingming Shu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hao Qu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Wei Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenhui Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qinghua Nie
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ruidong Xiang
- Agriculture Victoria, Agribio, Centre for AgriBiosciences, Bundoora, Victoria, Australia
- Cambridge-Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Shuli Liu
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Zhe Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhang Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA
| | - Hans Cheng
- Avian Disease and Oncology Laboratory, USDA, ARS, USNPRC, East Lansing, MI, USA
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Huaijun Zhou
- Department of Animal Science, University of California-Davis, Davis, CA, USA.
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark.
| |
Collapse
|
3
|
Sun J, Borowska D, Furniss JJ, Sutton K, Macqueen DJ, Vervelde L. Cellular landscape of avian intestinal organoids revealed by single cell transcriptomics. Sci Rep 2025; 15:11362. [PMID: 40175530 PMCID: PMC11965369 DOI: 10.1038/s41598-025-95721-4] [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: 01/30/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Studies of the avian gastrointestinal tract, where nutrient absorption and key host-pathogen interactions occur, have been strongly enabled by the development of intestinal organoid models. Here we report a single cell transcriptomic atlas of intestinal organoid cells derived from embryos of broiler and layer chickens, capturing mesenchymal, epithelial, endothelial, immune and neuronal cell lineages. Eight inferred mesenchymal subpopulations reflect anatomically distinct intestinal layers, including fibroblasts, telocytes, myofibroblasts, smooth myocytes, pericytes, and interstitial cells of Cajal. Identified heterogeneity within the epithelial lineage included enterocytes, goblet cells, Paneth cells, tuft cells, and diverse enteroendocrine cell subtypes. Additionally, we identified candidate macrophages, monocytes, γδ T cells, NK cells and granulocytes. Layer and broiler organoids showed significant differences in cell-specific transcriptome, most pronounced in epithelial cells, pointing to divergent selection on intestinal physiology. Our analysis finally provides a catalogue of novel cell marker genes to enable future research of chicken intestinal organoids.
Collapse
Affiliation(s)
- Jianxuan Sun
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Dominika Borowska
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - James J Furniss
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Kate Sutton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK.
| | - Lonneke Vervelde
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK.
- Royal GD, Deventer, The Netherlands.
| |
Collapse
|
4
|
Kaplan-Arabaci O, Dančišinová Z, Paulsen RE. The Chicken Embryo: An Alternative Animal Model in Development, Disease and Pharmacological Treatment. Pharmacol Res Perspect 2025; 13:e70086. [PMID: 40113588 PMCID: PMC11925699 DOI: 10.1002/prp2.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/14/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
To examine various medications and substances, in vivo models such as rats and mice are routinely used. However, it is utterly desirable to reduce extensive amounts of animals for these experimental models, which are costly and time-consuming. Animals are frequently put through a variety of procedures that could cause them pain, distress, or even harm; therefore, it is important to think about the ethical ramifications of using them in research. Thus, by following the three R's of animal research: reduction, replacement, and refinement, living animals used in studies should be minimized. The embryo of Gallus gallus, the domestic chicken, is a great model to research many different diseases and conditions. Its efficient blood supply from the chorioallantoic membrane gives us a unique possibility to administer chemicals or cells to the embryo in a noninvasive manner. In this review, we evaluate some advantages and disadvantages of using the developing chicken as an alternative in vivo model for development, disease, and pharmacological treatment. We focus on the top two leading causes of death: neurological disorders and cancer. We present a number of studies that describe the use of the chicken embryo in neuroscience and neurodevelopment research, in cancer research, and pharmacodynamic and pharmacokinetic studies. These studies show that the chicken embryo is an inexpensive, readily available, self-sufficient model with a short incubation period, high accessibility, and ideal for drug screening, making it an appealing model that can provide insightful biological and pharmacological information.
Collapse
Affiliation(s)
- Oykum Kaplan-Arabaci
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Zuzana Dančišinová
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovakia
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Campitelli LMM, Lopes KP, de Lima IL, Ferreira FB, Isidoro ND, Ferreira GM, Ponce MCF, Ferreira MCDO, Mendes LS, Marcelino PHR, Neves MM, Klein SG, Fonseca BB, Polveiro RC, da Silva MV. Methodological and Ethical Considerations in the Use of Chordate Embryos in Biomedical Research. Int J Mol Sci 2025; 26:2624. [PMID: 40141265 PMCID: PMC11941781 DOI: 10.3390/ijms26062624] [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: 01/10/2025] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Animal embryos are vital tools in scientific research, providing insights into biological processes and disease mechanisms. This paper explores their historical and contemporary significance, highlighting the shift towards the refinement of in vitro systems as alternatives to animal experimentation. We have conducted a data review of the relevant literature on the use of embryos in research and synthesized the data to highlight the importance of this model for scientific progress and the ethical considerations and regulations surrounding embryo research, emphasizing the importance of minimizing animal suffering while promoting scientific progress through the principles of replacement, reduction, and refinement. Embryos from a wide range of species, including mammals, fish, birds, amphibians, and reptiles, play a crucial experimental role in enabling us to understand factors such as substance toxicity, embryonic development, metabolic pathways, physiological processes, etc., that contribute to the advancement of the biological sciences. To apply this model effectively, it is essential to match the research objectives with the most appropriate methodology, ensuring that the chosen approach is appropriate for the scope of the study.
Collapse
Affiliation(s)
- Laura Maria Mendes Campitelli
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Karina Pereira Lopes
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Isabela Lemos de Lima
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Flávia Batista Ferreira
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Nayara Delfim Isidoro
- Faculty of Veterinary Medicine, Federal University of Uberlândia, Uberlândia 38410-337, MG, Brazil
| | - Giovana Magalhães Ferreira
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Maria Clara Fioravanti Ponce
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | | | - Ludmilla Silva Mendes
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Pedro Henrique Ribeiro Marcelino
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Matheus Morais Neves
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Sandra Gabriela Klein
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | | | - Richard Costa Polveiro
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
| | - Murilo Vieira da Silva
- Biotechnology in Experimental Models Laboratory—LABME, Federal University of Uberlândia, Uberlândia 38405-330, MG, Brazil; (L.M.M.C.); (M.M.N.)
- Rodent Animal Facilities Complex, Federal University of Uberlândia, Uberlândia 38400-902, MG, Brazil
| |
Collapse
|
6
|
Mon KKZ, Kenney LJ. Evaluation of tumor-colonizing Salmonella strains using the chick chorioallantoic membrane model. mBio 2025; 16:e0359024. [PMID: 39873483 PMCID: PMC11898558 DOI: 10.1128/mbio.03590-24] [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: 11/20/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
The chick embryo chorioallantoic membrane (CAM) tumor model is a valuable preclinical model for studying the tumor-colonizing process of Salmonella enterica serovar Typhimurium. It offers advantages such as cost-effectiveness, rapid turnaround, reduced engraftment issues, and ease of observation. In this study, we explored and validated the applicability of the partially immune-deficient CAM tumor model. Herein, we demonstrate that Salmonella preferentially colonizes tumors and directly causes tumor cell death. Bacterial migration, tumor colonization, and intra-tumor distribution did not require flagellar-mediated motility. The vast majority of Salmonella that colonized the CAM tumor were extracellular. Thus, tumor invasion was independent of both Salmonella pathogenicity island-1-encoded and Salmonella pathogenicity island-2-encoded type III secretion systems. Surprisingly, the extracellular residence of Salmonella on CAM tumors did not require biofilm formation. We evaluated our wild-type parental strain compared to the attenuated clinical strain VNP20009 and discovered a reduced tumor colonization capability of VNP20009. The inability to effectively colonize CAM tumors potentially explains the reduced anti-tumor efficacy of VNP20009. Our work establishes the xenograft CAM model as an informative and predictive screening platform for studying tumor-colonizing Salmonella.IMPORTANCECancer has a major impact on society, as it poses a significant health burden to human populations worldwide. Salmonella Typhimurium has demonstrated promise in cancer treatment by exerting direct tumoricidal effects and enhancing host-mediated anti-tumor immunity in xenograft mouse studies. A general understanding of its pathogenesis and the relative ease of genetic manipulation support the development of attenuated strains for therapeutic use. Alternative in ovo models, such as the chorioallantoic membrane tumor model, present a suitable screening platform to accelerate the development of therapeutic strains. It allows for rapid evaluation of Salmonella strains to assess their efficacy and potential as oncolytic agents. The present study establishes that the in ovo tumor model can be utilized as a preclinical tool for evaluating oncolytic Salmonella, bridging the gap between in vitro and in vivo screening.
Collapse
Affiliation(s)
- Khin K. Z. Mon
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Linda J. Kenney
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
7
|
Halabi S, Rocos N, Kaufman J. The search to understand the development of the chicken immune system: Differences in expression of MHC class I loci and waves of thymocytes as evolutionary relics? Dev Biol 2025; 519:38-45. [PMID: 39694171 DOI: 10.1016/j.ydbio.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/28/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Chickens are renowned as a model for embryogenesis but have also been responsible for crucial advances in virology, cancer research and immunology. However, chickens are best known as a major source of animal protein for human nutrition, with roughly 80 billion chickens alive each year supplying meat and eggs, the vast majority part of a global poultry industry. As a result, avian immunology been studied intensively for over 60 years, and it has become clear that a major genetic locus in chickens determining resistance to infectious disease and response to vaccines is the major histocompatibility complex (MHC). Compared to typical mammals, the chicken MHC is compact and simple, with only two classical class I genes. A dominantly-expressed class I gene, BF2, is the major ligand for cytotoxic T lymphocytes (CTLs), while the other locus, BF1, is much less well-expressed, lacking in some MHC haplotypes, and is a ligand for natural killer (NK) cells. Cell surface class I expression in neonatal chicks is far less than in adults, and one possibility is that BF2 is not well-expressed early in ontogeny. A precedent is found for amphibians: the single classical class I molecule is not expressed in tadpoles of Xenopus frogs, although non-polymorphic (and thus non-classical) class I molecules from the XNC locus are expressed, which are recognised for immune defence by non-canonical NKT lymphocytes. Indeed, three waves of different T cells are produced by the Xenopus thymus: in tadpoles, during metamorphosis and finally as adults. Three waves of thymic emigrants are also found for chickens, and reasoning by analogy, it may be that the waves of thymocytes and the expression of class I molecules during ontogeny of chickens are evolutionary relics. As well as scientific interest in the ontogeny of MHC class I expression and appearance of peripheral T cells, there are potential practical implications, given the importance of vaccination in ovo and in day-old chicks for the poultry industry.
Collapse
Affiliation(s)
- Samer Halabi
- University of Edinburgh, Institute for Immunology and Infection Research, Edinburgh, United Kingdom
| | - Nicolas Rocos
- University of Edinburgh, Institute for Immunology and Infection Research, Edinburgh, United Kingdom
| | - Jim Kaufman
- University of Edinburgh, Institute for Immunology and Infection Research, Edinburgh, United Kingdom.
| |
Collapse
|
8
|
Varela MF, Oria M, Poling HM, Lopriore E, Peiro JL. Development and Validation of a Minimally Invasive Transuterine Experimental Model of Gastroschisis. J Pediatr Surg 2025; 60:162163. [PMID: 39823694 DOI: 10.1016/j.jpedsurg.2025.162163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/13/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Perinatal management of gastroschisis remains a subject of substantial research. Current models, including teratogenic, genetic, and surgical approaches, often fail to accurately replicate gastroschisis, exhibiting limitations such as inaccurate phenotyping, low success rates, high mortality, lack of scientific validation, and significant technical challenges. Refined disease models are essential for improving the understanding of GS. This study seeks to develop and validate a minimally invasive transuterine experimental model of GS that overcomes these existing constraints to advance gastroschisis research. METHODS A gastroschisis model was surgically created in rat fetuses at E17 (n = 51 fetuses from n = 13 dams). Intestines were harvested at term and divided into herniated gastroschisis (GS-H), intra-abdominal gastroschisis (GS-I), and control (Co) groups. Morphometric analysis, histopathological examination, immunohistochemistry for interstitial cells of Cajal (ICC), double immunofluorescence for ICC and mast cells, TUNEL assay for apoptotic cells, and multiplex cytokine assay were performed to assess intestinal architecture, inflammation, ICC network, apoptosis, and cytokine levels across studied groups. RESULTS Histology from GS intestines revealed subchronic inflammation, peel formation, and architectural disruption. Herniated intestines exhibited a significantly increased weight/length ratio and thicker outer layers (p < 0.001) compared with control intestines. Herniated intestines had elevated inflammatory cytokine levels (GS-H vs GS-I and Co, p < 0.05 for G-CSF, GM-CSF, IL-12p70, IL-1beta) and increased apoptotic activity. CONCLUSIONS We developed and validated a new surgical model of GS that offers improved survival and feasibility. The key morphological changes and molecular markers observed in this experimental model resemble human gastroschisis.
Collapse
Affiliation(s)
- Maria Florencia Varela
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Neonatology, Department of Pediatrics, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Marc Oria
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Holly Marie Poling
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Jose Luis Peiro
- The Center for Fetal and Placental Research, Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH 45229, USA; University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH 45267, USA.
| |
Collapse
|
9
|
Mosna MJ, Garde FJ, Stinson MG, Pastore CD, Carcagno AL. The chorioallantoic membrane (CAM) model: From its origins in developmental biology to its role in cancer research. Dev Biol 2025; 519:79-95. [PMID: 39694172 DOI: 10.1016/j.ydbio.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/24/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Over the past century, the chick embryo model, historically employed for research in developmental biology, has become a valuable tool for cancer research. The characteristics of the chick chorioallantoic membrane (CAM) make it a convenient model for the study of cancer, leading to the establishment of the CAM assay as an alternative to traditional in vivo cancer models. In this review we will explore the characteristics of the CAM that make it suitable for cancer research, as well as its consolidation as a versatile platform in this field. We will put particular emphasis on describing the key features that make this model an important asset for studying the hallmarks of cancer and for testing a wide variety of therapeutic strategies for its treatment, and which make it a suitable host for patient-derived xenografts (PDX). Additionally, we will examine the wide spectrum of methodological approaches available to study these subjects, highlighting some innovative cases. Finally, we will discuss the advantages and disadvantages of the chick CAM as a model for cancer research and how we can improve this model to its full potential.
Collapse
Affiliation(s)
- María Jimena Mosna
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Federico J Garde
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Marcelo G Stinson
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Candela D Pastore
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Abel L Carcagno
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina.
| |
Collapse
|
10
|
Wagner BJ, Ettner-Sitter A, Ihlo NA, Behr M, Koelbl S, Brunner SM, Weber F, Rau BM, Schlitt HJ, Brochhausen C, Schoenmehl R, Artinger A, Schott D, Pizon M, Pachmann K, Aung T, Haerteis S, Hackl C. Patient-derived xenografts from circulating cancer stem cells as a preclinical model for personalized pancreatic cancer research. Sci Rep 2025; 15:2896. [PMID: 39843495 PMCID: PMC11754431 DOI: 10.1038/s41598-025-87054-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Patient-derived xenografts (PDXs) provide biologically relevant models and potential platforms for the development of treatment strategies for precision medicine in pancreatic cancer. Furthermore, circulating epithelial tumor cells (CETCs/CTCs) are released into the bloodstream by solid tumors and a rare subpopulation-circulating cancer stem cells (cCSCs) - is considered to be responsible for recurrence and plays a key role in metastasis. For the identification of cCSCs, an innovative in vitro assay to generate tumorspheres was established in this study. The number of tumorspheres and CETCs/CTCs was analyzed perioperatively in 25 pancreatic cancer patients. Additionally, an individual in vivo chorioallantoic membrane (CAM) culture system was used to generate PDXs from these tumorspheres. While overall correlations of CETCs/CTCs with clinicopathological parameters did not reach statistical significance, a significant difference in the number of tumorspheres was observed between patient subgroups with lower and higher UICC stages. This finding underscores their potential as biomarkers, providing valuable insights into clinical decision-making and tumor progression. The application of tumorspheres on the CAM successfully established PDXs within 7 days. These xenografts closely resembled the histological features of the primary tumor. Hence, this model represents a novel and fast option for individualized testing of new therapies for PDAC.
Collapse
Affiliation(s)
- Benedikt J Wagner
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Andreas Ettner-Sitter
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
| | - Nicolas A Ihlo
- Faculty of Informatics and Data Science, University of Regensburg, Bajuwarenstrasse 4, 93053, Regensburg, Germany
| | - Merle Behr
- Faculty of Informatics and Data Science, University of Regensburg, Bajuwarenstrasse 4, 93053, Regensburg, Germany
| | - Sebastian Koelbl
- Technology Campus Hutthurm, Deggendorf Institute of Technology, Hochleiten 1, 94116, Hutthurm, Germany
| | - Stefan M Brunner
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Bettina M Rau
- Department of General, Visceral and Thoracic Surgery, Academic Teaching Hospital Neumarkt, Nuernberger Strasse 12, 92318, Neumarkt in der Oberpfalz, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Rebecca Schoenmehl
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Annalena Artinger
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | | | - Monika Pizon
- Simfo GmbH, Kurpromenade 2, 95448, Bayreuth, Germany
- Labor Dr. Pachmann Bayreuth, Kurpromenade 2, 95448, Bayreuth, Germany
| | - Katharina Pachmann
- Simfo GmbH, Kurpromenade 2, 95448, Bayreuth, Germany
- Labor Dr. Pachmann Bayreuth, Kurpromenade 2, 95448, Bayreuth, Germany
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Dieter-Goerlitz-Platz 1, 94469, Deggendorf, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany.
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
11
|
Meijer MMY, van den Brand H, Niknafs S, Roura E. In ovo delivery of carvacrol triggers expression of chemotactic factors, antimicrobial peptides and pro-inflammatory pathways in the yolk sac of broiler chicken embryos. J Anim Sci Biotechnol 2025; 16:8. [PMID: 39828746 PMCID: PMC11742807 DOI: 10.1186/s40104-024-01131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Broiler chickens are most vulnerable immediately after hatching due to their immature immune systems, making them susceptible to infectious diseases. The yolk plays an important role in early immune defence by showing relevant antioxidant and passive immunity capabilities during broiler embryonic development. The immunomodulatory effects of phytogenic compound carvacrol have been widely reported. After in ovo delivery in the amniotic fluid during embryonic development carvacrol is known to migrate to the yolk sac. However, it is unknown whether carvacrol in the yolk could enhance defence responsiveness in the yolk sac. Therefore, the aim of this study was to improve early immune function in chicken embryos, and it was hypothesized that in ovo delivery of carvacrol would result in immunomodulatory effects in the yolk sac, potentially improving post-hatch resilience. METHODS On embryonic day (E)17.5, either a saline (control) or carvacrol solution was injected into the amniotic fluid. Yolk sac tissue samples were collected at E19.5, and transcriptomic analyses using RNA sequencing were performed, following functional enrichment analyses comparing the control (saline) and carvacrol-injected groups. RESULTS The results showed that 268 genes were upregulated and 174 downregulated in the carvacrol group compared to the control (P < 0.05; logFC < -0.5 or log FC > 0.5). Functional analyses of these differentially expressed genes, using KEGG, REACTOME, and Gene Ontology databases, showed enrichment of several immune-related pathways. This included the pathways 'Antimicrobial peptides' (P = 0.001) and 'Chemoattractant activity' (P = 0.004), amongst others. Moreover, the 'NOD-like receptor signaling' pathway was enriched (P = 0.002). Antimicrobial peptides are part of the innate immune defence and are amongst the molecules produced after the nucleotide oligomerization domain (NOD)-like receptor pathway activation. While these responses may be associated with an inflammatory reaction to an exogenous threat, they could also indicate that in ovo delivery of carvacrol could prepare the newly hatched chick against bacterial pathogens by potentially promoting antimicrobial peptide production through activation of NOD-like receptor signaling in the yolk sac. CONCLUSION In conclusion, these findings suggest that in ovo delivery of carvacrol has the potential to enhance anti-pathogenic and pro-inflammatory responses in the yolk sac via upregulation of antimicrobial peptides, and NOD-like receptor pathways.
Collapse
Affiliation(s)
- Mila M Y Meijer
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Henry van den Brand
- Department of Animal Sciences, Adaptation Physiology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Shahram Niknafs
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Eugeni Roura
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia.
| |
Collapse
|
12
|
Ayalew H, Xu C, Adane A, Sanchez ALB, Li S, Wang J, Wu S, Qiu K, Qi G, Zhang H. Ontogeny and function of the intestinal epithelial and innate immune cells during early development of chicks: to explore in ovo immunomodulatory nutrition. Poult Sci 2025; 104:104607. [PMID: 39693955 PMCID: PMC11720616 DOI: 10.1016/j.psj.2024.104607] [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: 09/17/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Intestinal epithelial cells (IECs) and innate immune cells in the gastrointestinal tract (GIT) of chickens play crucial roles in pathogens defense and maintaining gut health. However, their effectiveness influenced with their developmental and functional stages during pre and post hatch periods of chick. During embryonic development, differentiation and migration of these innate immune systems are tightly regulated by diverse cellular and molecular factors. The maturation and functionality of IECs are histologically evident starting embryonic day (ED) 14. Moreover, the innate immun cells, such as dendritic cells (DCs), macrophages, natural killer (NK) cells, and gamma-delta (γδ) T cells have showed developmental expression varation, while most identified by the 3rd days of incubation and capable of responsing to their cognate ligands of pathogens by ED 17, it may not efficient during posthatch period. In modern poultry production, in ovo feeding of bioactive substances is a topic of interest to maximize the protection capability of hatched chicks by enhancing improvement on the development of innate immune systems. However, their actions and effects on each distinct innate immune involved response are inconsistent and not clearly understood. Thus, summarizing the ontogeny and function of IECs, innate immunity systems, and interaction mechanisms of in ovo feeding of bioactive substances could provide baseline information for designing targeted in ovo feeding interventions to modulate cell waise specific innate immune systems.
Collapse
Affiliation(s)
- Habtamu Ayalew
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; University of Gondar, College of Veterinary Medicine and Animal Sciences, Po. Box 196, Gondar, Ethiopia
| | - Changchun Xu
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Assefa Adane
- University of Gondar, College of Veterinary Medicine and Animal Sciences, Po. Box 196, Gondar, Ethiopia
| | - Astrid Lissette Barreto Sanchez
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Siman Li
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jing Wang
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Shugeng Wu
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Kai Qiu
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Guanghai Qi
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haijun Zhang
- Laboratory of Quality and Safety Risk Assessment for Animal Products on Feed Hazards (Beijing) of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
13
|
Meijer MMY, Brand HVD, Palmieri C, Niknafs S, Khaskheli AA, Roura E. Early-life immunomodulation by carvacrol delivered in ovo in broiler chickens. Poult Sci 2024; 103:104286. [PMID: 39326180 PMCID: PMC11470485 DOI: 10.1016/j.psj.2024.104286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
In the first 2 wk after hatching, broiler chickens are vulnerable to enteric pathogens due to underdeveloped gastrointestinal and immune systems. Carvacrol has been reported to improve digestive and immune functions. This study aimed to optimize immune development of broiler chickens by delivering carvacrol in ovo. Effects of 2 in ovo treatments delivered at embryonic day (E)17.5 (saline or carvacrol) were evaluated at 3 stages (E19.5, hatch, and d 14 posthatch). Hatchability, performance parameters, lymphoid organ and yolk sac weights were determined. Histomorphology assessment was performed for jejunal samples at hatch and bursa of Fabricius samples at hatch and d 14. Gene expression of immune-relevant genes was determined for jejunal, bursal, and yolk sac samples over time. At hatch, BW was 0.85% lower (P = 0.02) after in ovo carvacrol delivery compared to the controls. Interactions between in ovo treatment and age were found for gene expression. At hatch, carvacrol treatment resulted in lower expression of proinflammatory cytokines IL-8 and IFN-γ in the yolk sac compared to the controls (P = 0.05 and < .001, respectively) suggesting a potential role for carvacrol-mediated immune modulation. At d 14, carvacrol treatment led to lower expression of proinflammatory cytokine IL-6 in the bursa compared to the controls (P = 0.002). In ovo carvacrol delivery led to bursal histomorphometric changes, including a larger cortex in the bursal follicles (P = 0.03), and a higher cortex/medulla ratio (P = 0.04) compared to the controls, indicating increased B-cell stimulation and maturation. Main effects were found for carvacrol treatment in the jejunum, with overall higher expression of proinflammatory mediators IL-1β and NF-κB, and anti-inflammatory IL-10 compared to the controls (P = 0.04, 0.02, and 0.02 respectively) from E19.5 to d 14. Age-related main effects showed various alterations in expression dynamics of immune-related genes across all tissues over time. Our findings suggest changes in immune parameters occur as the chicken develops, but these mostly do not interact with in ovo carvacrol treatment. In ovo carvacrol treatment alters immune activity of broiler chickens independent of age.
Collapse
Affiliation(s)
- Mila M Y Meijer
- Centre for Animal Science and Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Henry van den Brand
- Department of Animal Sciences, Adaptation Physiology Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Chiara Palmieri
- School of Veterinary Science, University of Queensland, Brisbane, Australia
| | - Shahram Niknafs
- Centre for Animal Science and Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Asad A Khaskheli
- Centre for Animal Science and Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia
| | - Eugeni Roura
- Centre for Animal Science and Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Brisbane, Australia.
| |
Collapse
|
14
|
Bodman-Harris O, Rollier CS, Iqbal M. Approaches to Enhance the Potency of Vaccines in Chickens. Vaccines (Basel) 2024; 12:1337. [PMID: 39771998 PMCID: PMC11680195 DOI: 10.3390/vaccines12121337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Outbreaks of avian pathogens such as Newcastle disease virus, avian influenza virus, and salmonella have a major impact on economies and food security worldwide. Some pathogens also pose a significant zoonotic potential, especially avian influenza viruses. Vaccination plays a key role in controlling many poultry diseases, and there are many vaccines licenced in the United Kingdom for diseases of poultry caused by viruses, bacteria, and parasites. However, these vaccines often do not provide complete protection and can cause unwanted side effects. Several factors affect the potency of poultry vaccines, including the type of vaccination used, the mechanism of delivery, and the use of adjuvants. Advancements in technology have led to the study and development of novel vaccines and vaccine adjuvants for use in poultry. These induce stronger immune responses compared with current vaccine technology and have the potential to protect against multiple poultry diseases. This review aims to discuss the existing poultry vaccine technology; the effect of delivery mechanisms on vaccine efficacy; the use of current and novel adjuvants; the ability to target antigens to antigen-presenting cells; and the use of probiotics, multivalent vaccines, and nanotechnology to enhance the potency of poultry vaccines.
Collapse
Affiliation(s)
- Oenone Bodman-Harris
- Avian Influenza and Newcastle Disease Research Group, The Pirbright Institute, Ash Road, Woking GU 24 0NF, UK;
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford GU2 7XH, UK;
| | - Christine S. Rollier
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford GU2 7XH, UK;
| | - Munir Iqbal
- Avian Influenza and Newcastle Disease Research Group, The Pirbright Institute, Ash Road, Woking GU 24 0NF, UK;
| |
Collapse
|
15
|
Zhang B, Gao K, Cao J, Xing G, Ji Z, Li Z, Li Y, Keijer J, Xie M, Zhou Z, Hou S, Tang J. Maternal riboflavin deficiency causes embryonic defects by activating ER stress-induced hepatocyte apoptosis pathway. Free Radic Biol Med 2024; 224:418-435. [PMID: 39241988 DOI: 10.1016/j.freeradbiomed.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Riboflavin deficiency (RD) induces liver damage, abnormal embryonic development, and high mortality. We hypothesized that the phenotype could be rescued by inhibiting ER stress. The objectives of the present study were to investigate the underlying molecular mechanisms of RD-induced embryonic defects using in vitro and in vivo models. Primary duck embryonic hepatocytes were treated with an ER stress inhibitor (4-PBA) or transfected with CHOP siRNA, and cultured in RD medium and riboflavin-sufficient (CON) medium for 8 days. Laying ducks (n = 20 cages/diet, 1 bird/cage) were fed an RD diet or CON diet for 14 wk, and the eggs were collected for hatching. At day 7 of incubation, the fertilized RD eggs were injected with or without 4-PBA into the yolk. RD decreased cell number and cell viability compared to the CON group, induced oxidative stress and apoptosis in primary duck embryonic hepatocytes. However, after being treated with an ER stress inhibitor (4-PBA) or transfected with CHOP siRNA, the apoptosis rate in RD hepatocytes decreased by 60.6 % and 86.1 %, respectively, being equal to the CON. These results indicated that RD-induced hepatocyte apoptosis is mediated by ER stress and the CHOP pathway. In vivo, RD embryos showed low hatchability, abnormal development, liver damage, ER stress, and apoptosis compared to the CON group. However, 4-PBA administration, as a model of ER stress inhibition, substantially restored embryonic development and alleviated liver damage in the RD group, including ER stress and apoptosis. Notably, hatchability in the RD group increased from 21.7 % to 72.7 % after 4-PBA treatment, though it remained less than the CON group (87.7 %). These results implicated ER stress-CHOP-apoptosis pathway as molecular mechanisms underlying RD-induced abnormal embryonic development and death, this target with potential for therapy or intervention.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Kexin Gao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Junting Cao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Guangnan Xing
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhanqing Ji
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhinan Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yating Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, 6700 AH Wageningen, the Netherlands
| | - Ming Xie
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhengkui Zhou
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shuisheng Hou
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Jing Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
16
|
Garcia P, Wang Y, Viallet J, Mehdi NEH, Montaut E, Decaens T, Emadali A, Macek Jílková Z. Liver cancer in ovo models for preclinical testing. FASEB J 2024; 38:e70029. [PMID: 39215630 DOI: 10.1096/fj.202401416r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapies have significantly improved the prognosis of patients with advanced hepatocellular carcinoma (HCC), although more than 70% of patients still do not respond to this first-line treatment. Many new combination strategies are currently being explored, which drastically increases the need for preclinical models that would allow large-scale testing of new immunotherapies and their combinations. We developed several in ovo (in the egg) human liver cancer models, based on human tumor xenografts of different liver cancer cell lines on the chicken embryo's chorioallantoic membrane. We characterized the angiogenesis, as well as the collagen accumulation and tumor immune microenvironment, and tested atezolizumab (anti-PD-L1) plus bevacizumab (anti-VEGF) treatment. Our results show the involvement of chicken immune cells in tumor growth, reproducing a classical non-inflamed "cold" as well as inflamed "hot" tumor status, depending on the in ovo liver cancer model. The treatment by atezolizumab and bevacizumab was highly efficient in the "hot" tumor model PLC/PRF/5 in ovo with the reduction of tumor size by 76% (p ≤ .0001) compared with the control, whereas the efficacy was limited in the "cold" Hep3B in ovo tumor. The contribution of the anti-PD-L1 blockade to the anti-tumoral effect in the PLC/PRF/5 in ovo model was demonstrated by the efficacy of atezolizumab monotherapy (p = .0080, compared with the control). To conclude, our study provides a detailed characterization and rational arguments that could help to partially replace conventional laboratory animals with a more ethical model, suited to the current needs of preclinical research of new immunotherapies for liver cancer.
Collapse
Affiliation(s)
- Paul Garcia
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- R&D Department, Inovotion, La Tronche, France
| | - Yan Wang
- R&D Department, Inovotion, La Tronche, France
| | | | - Nour El Houda Mehdi
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Emilie Montaut
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Pôle Recherche, CHU Grenoble Alpes, La Tronche, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Service d'Hépato-gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, La Tronche, France
| | - Anouk Emadali
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Pôle Recherche, CHU Grenoble Alpes, La Tronche, France
| | - Zuzana Macek Jílková
- Univ. Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Service d'Hépato-gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, La Tronche, France
| |
Collapse
|
17
|
Das J, Roy Sarkar S, Das A, Barui A, Mitra Mazumder P. Assessment of efficacy of chrysin in diabetes-associated cardiac complications in chick embryo and murine model. J Pharm Pharmacol 2024; 76:1225-1235. [PMID: 38989974 DOI: 10.1093/jpp/rgae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVES Patients with type 2 diabetes or prolonged diabetic condition are webbed into cardiac complications. This study aimed to ascertain the utility of chick embryo as an alternative to the mammalian model for type 2 diabetes-induced cardiac complications and chrysin as a protective agent. METHODS Diabetes was activated in ovo model (chick embryo) using glucose along with β-hydroxybutyric acid. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Alamar, and Kenacid blue assay were used to compare with chrysin-administered group. Blood glucose level, total cholesterol, triglyceride, and high-density lipoprotein were considered as endpoints. Diabetes was induced in Wistar albino rats by administering a high-fat diet and a subdued dose of streptozotocin (35 mg/kg, b.w). Percentage of glycated hemoglobin, creatinine kinase-MB, tumor necrosis factor-α, and C-reactive protein were evaluated and compared with chrysin administered group. KEY FINDINGS Chrysin treatment improved elevated blood glucose levels and dyslipidemia in a diabetic group of whole embryos. Condensed cellular growth and protein content as well as enhanced cytotoxicity in ovo were shielded by chrysin. Chrysin reduced cardiac and inflammatory markers in diabetic rats and provided cellular protection to damage the heart of diabetic rats. CONCLUSION The protective action of chrysin in ovo model induced a secondary complication associated with diabetes, evidenced that the ovo model is an effective alternative in curtailing higher animal use in scientific research.
Collapse
Affiliation(s)
- Joyani Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Suparna Roy Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Ankita Das
- Centre for Health Care Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur 711103, India
| | - Ananya Barui
- Centre for Health Care Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur 711103, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, India
| |
Collapse
|
18
|
Wang Y, Zou M, Liu J, Guo Q, Lv S, Chen C, Wang T, Zhao W, Li S, Peng X. Alarming and calming: Dual functions of S100A9 on Mycoplasma gallisepticun infection in avian cells. Vet Microbiol 2024; 296:110175. [PMID: 39018941 DOI: 10.1016/j.vetmic.2024.110175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/09/2024] [Accepted: 07/04/2024] [Indexed: 07/19/2024]
Abstract
Mycoplasma gallisepticum (MG) is the primary causative agent of chronic respiratory disease (CRD) in chickens, characterized by respiratory inflammation. S100A9 plays a pivotal role in modulating the inflammatory response to microbial pathogens. Our prior investigation revealed a significant upregulation of S100A9 in the lungs of chickens following MG infection. This study delves into the immunomodulatory effects of S100A9 during MG infection, demonstrating a notable increase in S100A9 levels in the lungs, immune organs, alveolar epithelial type II cells (AECII), and macrophage HD11 cells of MG-infected chicks and embryos. In MG-infected AECII cells, S100A9 overexpression significantly enhanced MG proliferation and adhesion, suppressed AVBD1, NFκB, pro-inflammatory factors (IL1β and TNFα), and chemokines, reduced apoptosis, and promoted cell proliferation, thereby facilitating MG infection. Conversely, inhibiting S100A9 produced opposing effects. In MG-infected HD11 cells, S100A9 impeded MG proliferation and adhesion, increased AVBD1, NFκB, pro-inflammatory factors, and chemokines, and induced cell apoptosis while inhibiting proliferation. Additional results demonstrated that S100A9 facilitates MG infection by modulating the TLR7/NFκB/JAK/STAT pathway in AECII/HD11 cells. In summary, S100A9 exhibits a dual role in activating/inhibiting the natural immune response through TLR7/NFκB/JAK/STAT pathway regulation. This dual role promotes MG infection in AECII cells while enabling MG to evade immune surveillance by HD11 cells, ultimately enhancing the overall infection process. These findings advance our understanding of host-pathogen interactions during MG infection and underscore S100A9's potential as a therapeutic target for CRD in chickens.
Collapse
Affiliation(s)
- Yingjie Wang
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Mengyun Zou
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Jin Liu
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiao Guo
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Shan Lv
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Chunyu Chen
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Tengfei Wang
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenqing Zhao
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Shiying Li
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuli Peng
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
19
|
Ceccopieri C, Madej JP. Chicken Secondary Lymphoid Tissues-Structure and Relevance in Immunological Research. Animals (Basel) 2024; 14:2439. [PMID: 39199973 PMCID: PMC11350708 DOI: 10.3390/ani14162439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Recent discoveries have indicated the importance of developing modern strategies for vaccinations, more ethical research models, and effective alternatives to antibiotic treatment in farm animals. Chickens (Gallus gallus) play a crucial role in this context given the commercial and economic relevance of poultry production worldwide and the search for analogies between the immune systems of humans and birds. Specifically, chicken secondary lymphoid tissues share similar features to their human counterparts. Chickens have several secondary or peripheral lymphoid tissues that are the sites where the adaptive immune response is initiated. The more general classification of these organs divides them into the spleen and skin-, pineal-, or mucosa-associated lymphoid tissues. Each of these tissues is further subdivided into separate lymphoid structures that perform specific and different functions along the animal's body. A review summarizing the state of the art of research on chicken secondary lymphoid organs is of great relevance for the design of future studies.
Collapse
Affiliation(s)
| | - Jan P. Madej
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| |
Collapse
|
20
|
Song C, Xiong DD, He RQ, Yong XZ, Huang ZG, Dang YW, Chen G, Pang YY, Zhao CY, Qu N, Wei DM. Bibliometric study of the application of the chicken embryo chorioallantoic membrane model in cancer research: the top 100 most cited articles. J Comp Pathol 2024; 213:59-72. [PMID: 39116802 DOI: 10.1016/j.jcpa.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/26/2024] [Accepted: 07/07/2024] [Indexed: 08/10/2024]
Abstract
The chicken embryo chorioallantoic membrane (CAM) model has played a crucial role in various aspects of cancer research. The purpose of this study is to help researchers clarify the research direction and prospects of the CAM model. A bibliometric analysis was conducted on the top 100 most cited articles on use of the CAM model in tumour research, retrieved from the Web of Science Core Collection database. Tools such as Bibliometrix, VOSviewer, CiteSpace and Excel were utilized for the visualization network analysis. The 100 articles analysed were mainly from the USA, China and European countries such as Germany and France. Tumour research involving CAM model experiments demonstrated reliability and scientific rigor (average citation count = 156.2). The analysis of keywords, topics and subject areas revealed that the applications of this model ranged from the biological characteristics of tumours to molecular mechanisms and signaling pathways, to recent developments in nanotechnology and clinical applications. Additionally, nude mouse experiments have been more frequently performed in recent years. We conclude that the CAM model is efficient, simple and cost-effective, and has irreplaceable value in various aspects of cancer research. In the future, the CAM model can further contribute to nanotechnology research.
Collapse
Affiliation(s)
- Chang Song
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Dan-Dan Xiong
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiang-Zhi Yong
- Department of Periodontal and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yu-Yan Pang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chun-Yan Zhao
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Ning Qu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Dan-Ming Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
21
|
de Souza JB, Sommerfeld S, Almeida-Souza HO, Vaz ER, Bastos LM, Santos FDAA, Rodrigues AC, Medeiros-Ronchi AA, Goulart LR, Fonseca BB. A new standardization for the use of chicken embryo: selection of target from the phage display library and infection. Appl Microbiol Biotechnol 2024; 108:412. [PMID: 38985354 PMCID: PMC11236870 DOI: 10.1007/s00253-024-13227-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024]
Abstract
The filamentous bacteriophage M13KO7 (M13) is the most used in phage display (PD) technology and, like other phages, has been applied in several areas of medicine, agriculture, and in the food industry. One of the advantages is that they can modulate the immune response in the presence of pathogenic microorganisms, such as bacteria and viruses. This study evaluated the use of phage M13 in the chicken embryos model. We inoculated 13-day-old chicken embryos with Salmonella Pullorum (SP) and then evaluated survival for the presence of phage M13 or E. coli ER2738 (ECR) infected with M13. We found that the ECR bacterium inhibits SP multiplication in 0.32 (M13-infected ECR) or 0.44 log UFC/mL (M13-uninfected ECR) and that the ECR-free phage M13 from the PD library can be used in chicken embryo models. This work provides the use of the chicken embryo as a model to study systemic infection and can be employed as an analysis tool for various peptides that M13 can express from PD selection. KEY POINTS: • SP-infected chicken embryo can be a helpful model of systemic infection for different tests. • Phage M13 does not lead to embryonic mortality or cause serious injury to embryos. • Phage M13 from the PD library can be used in chicken embryo model tests.
Collapse
Affiliation(s)
- Jessica Brito de Souza
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Simone Sommerfeld
- Postgraduate Program in Veterinary Sciences, Faculty of Veterinary Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| | - Hebréia Oliveira Almeida-Souza
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil.
| | - Emília Rezende Vaz
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Luciana Machado Bastos
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Fabiana de Almeida Araújo Santos
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
- Postgraduate Program in Veterinary Sciences, Faculty of Veterinary Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| | - Alessandra Castro Rodrigues
- Postgraduate Program in Veterinary Sciences, Faculty of Veterinary Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Luiz Ricardo Goulart
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Belchiolina Beatriz Fonseca
- Postgraduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil.
- Postgraduate Program in Veterinary Sciences, Faculty of Veterinary Medicine, Federal University of Uberlândia, Uberlândia, Brazil.
| |
Collapse
|
22
|
Zhu Z, Hu Z, Ojima S, Yu X, Sugiyama M, Ono HK, Hu DL. Critical Involvement of the Thioredoxin Reductase Gene ( trxB) in Salmonella Gallinarum-Induced Systemic Infection in Chickens. Microorganisms 2024; 12:1180. [PMID: 38930562 PMCID: PMC11205728 DOI: 10.3390/microorganisms12061180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Salmonella enterica serovar Gallinarum biovar Gallinarum (SG) causes fowl typhoid, a notifiable infectious disease in poultry. However, the pathogenic mechanism of SG-induced systemic infection in chickens remains unclear. Thioredoxin reductase (TrxB) is a redox protein crucial for regulating various enzyme activities in Salmonella serovar, but the role in SG-induced chicken systemic infection has yet to be determined. Here, we constructed a mutant SG strain lacking the trxB gene (trxB::Cm) and used chicken embryo inoculation and chicken oral infection to investigate the role of trxB gene in the pathogenicity of SG. Our results showed that trxB::Cm exhibited no apparent differences in colony morphology and growth conditions but exhibited reduced tolerance to H2O2 and increased resistance to bile acids. In the chicken embryo inoculation model, there was no significant difference in the pathogenicity of trxB::Cm and wild-type (WT) strains. In the chicken oral infection, the WT-infected group exhibited typical clinical symptoms of fowl typhoid, with complete mortality between days 6 and 9 post infection. In contrast, the trxB::Cm group showed a 100% survival rate, with no apparent clinical symptoms or pathological changes observed. The viable bacterial counts in the liver and spleen of the trxB::Cm-infected group were significantly reduced, accompanied by decreased expression of cytokines and chemokines (IL-1β, IL-6, IL-12, CXCLi1, TNF-α, and IFN-γ), which were significantly lower than those in the WT group. These results show that the pathogenicity of the trxB-deficient strain was significantly attenuated, indicating that the trxB gene is a crucial virulence factor in SG-induced systemic infection in chickens, suggesting that trxB may become a potentially effective target for controlling and preventing SG infection in chickens.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
| | - Shinjiro Ojima
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Xiaoying Yu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (S.O.); (X.Y.); (H.K.O.)
| |
Collapse
|
23
|
Hu J, Zhang Z, Cai J, Hao S, Li C, Feng X. The Functional Mechanism of BP9 in Promoting B Cell Differentiation and Inducing Antigen Presentation. Vaccines (Basel) 2024; 12:607. [PMID: 38932336 PMCID: PMC11209236 DOI: 10.3390/vaccines12060607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
The Bursa of Fabricius, an avian unique humoral immune organ, is instrumental to B cell development. Bursal-derived peptide BP9 fosters B-cell development and formation. Yet, the exact mechanism wherein BP9 impacts B cell differentiation and antigenic presentation remains undefined. In this paper, B cell activation and differentiation in the spleen cells from mice immunized with the AIV vaccine and BP9 were detected following flow cytometry (FCM) analysis. Furthermore, the molecular mechanism of BP9 in B cell differentiation in vivo was investigated with RNA sequencing technology. To verify the potential functional mechanism of BP9 in the antigenic presentation process, the transcriptome molecular basis of chicken macrophages stimulated by BP9 was measured via high-throughput sequencing technology. The results proved that when given in experimental dosages, BP9 notably accelerated total B cells, and enhanced B-cell differentiation and plasma cell production. The gene expression profiles of B cells from mice immunized with 0.01 mg/mL BP9 and AIV vaccine disclosed that 0.01 mg/mL BP9 initiated the enrichment of several biological functions and significantly stimulated key B-cell pathways in immunized mice. Crucially, a total of 4093 differentially expressed genes were identified in B cells with BP9 stimulation, including 943 upregulated genes and 3150 downregulated genes. Additionally, BP9 induced various cytokine productions in the chicken macrophage HD11 cells and activated 9 upregulated and 20 downregulated differential miRNAs, which were involved in various signal and biological processes. Furthermore, BP9 stimulated the activation of multiple transcription factors in HD11 cells, which was related to antigen presentation processes. In summary, these results suggested that BP9 might promote B cell differentiation and induce antigen presentation, which might provide the valuable insights into the mechanism of B cell differentiation upon bursal-derived immunomodulating peptide stimulation and provide a solid experimental groundwork for enhancing vaccine-induced immunity.
Collapse
Affiliation(s)
- Jianing Hu
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ze Zhang
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiaxi Cai
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shanshan Hao
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chenfei Li
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiuli Feng
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.H.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
24
|
Bender L, Preis E, Engelhardt KH, Amin MU, Ayoub AM, Librizzi D, Roschenko V, Schulze J, Yousefi BH, Schaefer J, Bakowsky U. In vitro and in ovo photodynamic efficacy of nebulized curcumin-loaded tetraether lipid liposomes prepared by DC as stable drug delivery system. Eur J Pharm Sci 2024; 196:106748. [PMID: 38471594 DOI: 10.1016/j.ejps.2024.106748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Lung cancer is one of the most common causes of high mortality worldwide. Current treatment strategies, e.g., surgery, radiotherapy, chemotherapy, and immunotherapy, insufficiently affect the overall outcome. In this study, we used curcumin as a natural photosensitizer in photodynamic therapy and encapsulated it in liposomes consisting of stabilizing tetraether lipids aiming for a pulmonary drug delivery system against lung cancer. The liposomes with either hydrolyzed glycerol-dialkyl-glycerol tetraether (hGDGT) in different ratios or hydrolyzed glycerol-dialkyl-nonitol tetraether (hGDNT) were prepared by dual centrifugation (DC), an innovative method for liposome preparation. The liposomes' physicochemical characteristics before and after nebulization and other nebulization characteristics confirmed their suitability. Morphological characterization using atomic force and transmission electron microscopy showed proper vesicular structures indicative of liposomes. Qualitative and quantitative uptake of the curcumin-loaded liposomes in lung adenocarcinoma (A549) cells was visualized and proven. Phototoxic effects of the liposomes were detected on A549 cells, showing decreased cell viability. The generation of reactive oxygen species required for PDT and disruption of mitochondrial membrane potential were confirmed. Moreover, the chorioallantoic membrane (CAM) model was used to further evaluate biocompatibility and photodynamic efficacy in a 3D cell culture context. Photodynamic efficacy was assessed by PET/CT after nebulization of the liposomes onto the xenografted tumors on the CAM with subsequent irradiation. The physicochemical properties and the efficacy of tetraether lipid liposomes encapsulating curcumin, especially liposomes containing hGDNT, in 2D and 3D cell cultures seem promising for future PDT usage against lung cancer.
Collapse
Affiliation(s)
- Lena Bender
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Konrad H Engelhardt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Abdallah M Ayoub
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Damiano Librizzi
- Center for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging, Department of Nuclear Medicine, University of Marburg, Hans-Meerwein-Str. 3, Marburg 35043, Germany
| | - Valeri Roschenko
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Jan Schulze
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Behrooz H Yousefi
- Center for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging, Department of Nuclear Medicine, University of Marburg, Hans-Meerwein-Str. 3, Marburg 35043, Germany
| | - Jens Schaefer
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, Marburg 35037, Germany.
| |
Collapse
|
25
|
Bender L, Ayoub AM, Schulze J, Amin MU, Librizzi D, Engelhardt KH, Roschenko V, Yousefi BH, Schäfer J, Preis E, Bakowsky U. Evaluating the photodynamic efficacy of nebulized curcumin-loaded liposomes prepared by thin-film hydration and dual centrifugation: In vitro and in ovo studies. BIOMATERIALS ADVANCES 2024; 159:213823. [PMID: 38460353 DOI: 10.1016/j.bioadv.2024.213823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Lung cancer, one of the most common causes of high mortality worldwide, still lacks appropriate and convenient treatment options. Photodynamic therapy (PDT) has shown promising results against cancer, especially in recent years. However, pulmonary drug delivery of the predominantly hydrophobic photosensitizers still represents a significant obstacle. Nebulizing DPPC/Cholesterol liposomes loaded with the photosensitizer curcumin via a vibrating mesh nebulizer might overcome current restrictions. In this study, the liposomes were prepared by conventional thin-film hydration and two other methods based on dual centrifugation. The liposomes' physicochemical properties were determined before and after nebulization, showing that liposomes do not undergo any changes. However, morphological characterization of the differently prepared liposomes revealed structural differences between the methods in terms of lamellarity. Internalization of curcumin in lung adenocarcinoma (A549) cells was visualized and quantified. The generation of reactive oxygen species because of the photoreaction was also proven. The photodynamic efficacy of the liposomal formulations was tested against A549 cells. They revealed different phototoxic responses at different radiant exposures. Furthermore, the photodynamic efficacy was investigated after nebulizing curcumin-loaded liposomes onto xenografted tumors on the CAM, followed by irradiation, and evaluated using positron emission tomography/computed tomography and histological analysis. A decrease in tumor metabolism could be observed. Based on the efficacy of curcumin-loaded liposomes in 2D and 3D models, liposomes, especially with prior film formation, can be considered a promising approach for PDT against lung cancer.
Collapse
Affiliation(s)
- Lena Bender
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Abdallah M Ayoub
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Jan Schulze
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Damiano Librizzi
- Center for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging, Department of Nuclear Medicine, University of Marburg, Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| | - Konrad H Engelhardt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Valeri Roschenko
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Behrooz H Yousefi
- Center for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging, Department of Nuclear Medicine, University of Marburg, Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| | - Jens Schäfer
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| |
Collapse
|
26
|
Conci C, Sironi L, Jacchetti E, Panzeri D, Inverso D, Martínez Vázquez R, Osellame R, Collini M, Cerullo G, Chirico G, Raimondi MT. In vivo label-free tissue histology through a microstructured imaging window. APL Bioeng 2024; 8:016102. [PMID: 38222895 PMCID: PMC10787586 DOI: 10.1063/5.0165411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/06/2023] [Indexed: 01/16/2024] Open
Abstract
Tissue histopathology, based on hematoxylin and eosin (H&E) staining of thin tissue slices, is the gold standard for the evaluation of the immune reaction to the implant of a biomaterial. It is based on lengthy and costly procedures that do not allow longitudinal studies. The use of non-linear excitation microscopy in vivo, largely label-free, has the potential to overcome these limitations. With this purpose, we develop and validate an implantable microstructured device for the non-linear excitation microscopy assessment of the immune reaction to an implanted biomaterial label-free. The microstructured device, shaped as a matrix of regular 3D lattices, is obtained by two-photon laser polymerization. It is subsequently implanted in the chorioallantoic membrane (CAM) of embryonated chicken eggs for 7 days to act as an intrinsic 3D reference frame for cell counting and identification. The histological analysis based on H&E images of the tissue sections sampled around the implanted microstructures is compared to non-linear excitation and confocal images to build a cell atlas that correlates the histological observations to the label-free images. In this way, we can quantify the number of cells recruited in the tissue reconstituted in the microstructures and identify granulocytes on label-free images within and outside the microstructures. Collagen and microvessels are also identified by means of second-harmonic generation and autofluorescence imaging. The analysis indicates that the tissue reaction to implanted microstructures is like the one typical of CAM healing after injury, without a massive foreign body reaction. This opens the path to the use of similar microstructures coupled to a biomaterial, to image in vivo the regenerating interface between a tissue and a biomaterial with label-free non-linear excitation microscopy. This promises to be a transformative approach, alternative to conventional histopathology, for the bioengineering and the validation of biomaterials in in vivo longitudinal studies.
Collapse
Affiliation(s)
- Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Laura Sironi
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Davide Panzeri
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Maddalena Collini
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Giuseppe Chirico
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| |
Collapse
|
27
|
Pogorzelska A, Kuropka P, Stygar D, Michalczyk K, Chełmecka E, Zalejska-Fiolka J, Kublicka A, Janicka P, Bażanów B. Impact of Human Adenovirus 36 on Embryonated Chicken Eggs: Insights into Growth Mechanisms. Int J Mol Sci 2024; 25:2341. [PMID: 38397017 PMCID: PMC10889315 DOI: 10.3390/ijms25042341] [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: 12/20/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Human adenovirus 36 (HAdV-D36) is presently the sole virus identified to be associated with an elevated risk of obesity in both humans and animals. However, its impact on embryonated chicken eggs (ECEs) remains unexplored. This study endeavoured to examine the influence of HAdV-D36 on embryonic development by utilizing embryonated chicken eggs as a dynamic model. To simulate various infection routes, the allantoic cavity and the yolk sac of ECEs were inoculated with HAdV-D36. Subsequently, embryos from both the experimental (inoculated with virus) and control (inoculated with PBS) groups were weighed and subjected to daily histological examination. The daily embryo weights were assessed and compared between groups using the Shapiro-Wilk test. Histopathological changes in tissues were examined and compared between the tested and control groups to ascertain physiological alterations induced by the virus. Our study confirmed a significant increase in the body weight of ECEs. However, this phenomenon was not attributable to adipose tissue development; rather, it was characterized by an augmented number of cells in all observed tissues compared to control subjects. We posit that HAdV-D36 may impact developing organisms through mechanisms other than enhanced adipose tissue development. Specifically, our findings indicate an increased number of cells in all tissues, a phenomenon that occurs through an as-yet-unexplored pathway.
Collapse
Affiliation(s)
- Aleksandra Pogorzelska
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 31 C. K. Norwida Street, 50-573 Wroclaw, Poland; (A.P.); (A.K.); (P.J.)
| | - Piotr Kuropka
- Department of Biostructure and Animal Physiology, Division of Histology and Embryology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 25, 50-635 Wroclaw, Poland;
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Science in Zabrze, Medical University of Silesia, 19 Jordana Street, 40-808 Zabrze, Poland; (D.S.); (K.M.)
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Science in Zabrze, Medical University of Silesia, 19 Jordana Street, 40-808 Zabrze, Poland; (D.S.); (K.M.)
| | - Elżbieta Chełmecka
- Department of Medical Statistic, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 30 Ostrogórska Street, 40-200 Sosnowiec, Poland;
| | - Jolanta Zalejska-Fiolka
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Street, 41-808 Zabrze, Poland;
| | - Agata Kublicka
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 31 C. K. Norwida Street, 50-573 Wroclaw, Poland; (A.P.); (A.K.); (P.J.)
| | - Paulina Janicka
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 31 C. K. Norwida Street, 50-573 Wroclaw, Poland; (A.P.); (A.K.); (P.J.)
| | - Barbara Bażanów
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 31 C. K. Norwida Street, 50-573 Wroclaw, Poland; (A.P.); (A.K.); (P.J.)
| |
Collapse
|
28
|
Gautam H, Ayalew LE, Shaik NA, Subhasinghe I, Popowich S, Chow-Lockerbie B, Dixon A, Ahmed KA, Tikoo SK, Gomis S. Exploring the predictive power of jejunal microbiome composition in clinical and subclinical necrotic enteritis caused by Clostridium perfringens: insights from a broiler chicken model. J Transl Med 2024; 22:80. [PMID: 38243294 PMCID: PMC10799374 DOI: 10.1186/s12967-023-04728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/13/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Necrotic enteritis (NE) is a severe intestinal infection that affects both humans and poultry. It is caused by the bacterium Clostridium perfringens (CP), but the precise mechanisms underlying the disease pathogenesis remain elusive. This study aims to develop an NE broiler chicken model, explore the impact of the microbiome on NE pathogenesis, and study the virulence of CP isolates with different toxin gene combinations. METHODS This study established an animal disease model for NE in broiler chickens. The methodology encompassed inducing abrupt protein changes and immunosuppression in the first experiment, and in the second, challenging chickens with CP isolates containing various toxin genes. NE was evaluated through gross and histopathological scoring of the jejunum. Subsequently, jejunal contents were collected from these birds for microbiome analysis via 16S rRNA amplicon sequencing, followed by sequence analysis to investigate microbial diversity and abundance, employing different bioinformatic approaches. RESULTS Our findings reveal that CP infection, combined with an abrupt increase in dietary protein concentration and/or infection with the immunosuppressive variant infectious bursal disease virus (vIBDV), predisposed birds to NE development. We observed a significant decrease (p < 0.0001) in the abundance of Lactobacillus and Romboutsia genera in the jejunum, accompanied by a notable increase (p < 0.0001) in Clostridium and Escherichia. Jejunal microbial dysbiosis and severe NE lesions were particularly evident in birds infected with CP isolates containing cpa, netB, tpeL, and cpb2 toxin genes, compared to CP isolates with other toxin gene combinations. Notably, birds that did not develop clinical or subclinical NE following CP infection exhibited a significantly higher (p < 0.0001) level of Romboutsia. These findings shed light on the complex interplay between CP infection, the gut microbiome, and NE pathogenesis in broiler chickens. CONCLUSION Our study establishes that dysbiosis within the jejunal microbiome serves as a reliable biomarker for detecting subclinical and clinical NE in broiler chicken models. Additionally, we identify the potential of the genera Romboutsia and Lactobacillus as promising candidates for probiotic development, offering effective alternatives to antibiotics in NE prevention and control.
Collapse
Affiliation(s)
- Hemlata Gautam
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Lisanework E Ayalew
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Noor Ahmad Shaik
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Iresha Subhasinghe
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Shelly Popowich
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Betty Chow-Lockerbie
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Alexa Dixon
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Khawaja Ashfaque Ahmed
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Suresh K Tikoo
- Vaccinology and Immunotherapy, School of Public Health, University of Saskatchewan, 5D40 Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
29
|
Fraga LR, de Oliveira MR, Wermann KM, Vargesson N. Assessment of the Teratogenic Effect of Drugs on the Chicken Embryo. Methods Mol Biol 2024; 2753:251-260. [PMID: 38285343 DOI: 10.1007/978-1-0716-3625-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Pre-clinical trials are an essential step that underpins the drug discovery, development, and safety process. During this process, animal testing is performed to determine the safety of new compounds and any potential adverse effects. Developmental toxicity tests are carried out to verify whether the drug has potential to cause congenital anomalies to the developing embryo/fetus. Chicken embryos are very useful for these purposes and present several advantages, such as low cost of production and housing, easy handling and manipulation, and rapid development in addition to sharing similarities to the human embryo at molecular, cellular, and anatomical levels. In this chapter, we bring methods for using the chicken embryo model for testing the teratogenic effects of drugs and assessing the main outcomes of them.
Collapse
Affiliation(s)
- Lucas Rosa Fraga
- Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Maikel Rosa de Oliveira
- Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Karina Maria Wermann
- Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Neil Vargesson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
30
|
Li X, Liu X, Cui L, Liu Z, Zhang Y, Li H. How to Break through the Bottlenecks of in Ovo Vaccination in Poultry Farming. Vaccines (Basel) 2023; 12:48. [PMID: 38250861 PMCID: PMC10821430 DOI: 10.3390/vaccines12010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/15/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Poultry farming is one of the pillar industries of global animal husbandry. In order to guarantee production, poultry are frequently vaccinated from the moment they are hatched. Even so, the initial immunity of chicks is still very poor as they are in the "window period" of immune protection. In ovo vaccination pushes the initial immunization time forward to the incubation period, thereby providing earlier immune protection for chicks. In ovo vaccination is currently a research hotspot of poultry disease prevention and control, which is in line with the intensification of poultry production. However, the vaccines currently available for in ovo vaccination are limited and cannot meet the needs of industrial development, so how to efficiently activate the adaptive immune response of chicken embryos becomes the key to restrict product development and technological progress of in ovo vaccination. Its breakthrough, to a large extent, depends on systematic illustration of the mechanism underlying the adaptive immune response post immunization. Clarification of this issue will provide us with theoretical support and potential solutions for the development of novel vaccines for in ovo vaccination, the augmentation of efficacy of current vaccines and the optimization of immune programs.
Collapse
Affiliation(s)
- Xuefeng Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (X.L.); (X.L.)
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an 710061, China
| | - Xiaoxiao Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (X.L.); (X.L.)
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an 710061, China
| | - Lu Cui
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (L.C.); (Z.L.)
| | - Zheyi Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (L.C.); (Z.L.)
| | - Yu Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (X.L.); (X.L.)
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an 710061, China
| | - Hai Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (X.L.); (X.L.)
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an 710061, China
| |
Collapse
|
31
|
Schulze J, Schöne L, Ayoub AM, Librizzi D, Amin MU, Engelhardt K, Yousefi BH, Bender L, Schaefer J, Preis E, Schulz-Siegmund M, Wölk C, Bakowsky U. Modern Photodynamic Glioblastoma Therapy Using Curcumin- or Parietin-Loaded Lipid Nanoparticles in a CAM Model Study. ACS APPLIED BIO MATERIALS 2023; 6:5502-5514. [PMID: 38016693 PMCID: PMC10732153 DOI: 10.1021/acsabm.3c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Natural photosensitizers, such as curcumin or parietin, play a vital role in photodynamic therapy (PDT), causing a light-mediated reaction that kills cancer cells. PDT is a promising treatment option for glioblastoma, especially when combined with nanoscale drug delivery systems. The curcumin- or parietin-loaded lipid nanoparticles were prepared via dual asymmetric centrifugation and subsequently characterized through physicochemical analyses including dynamic light scattering, laser Doppler velocimetry, and atomic force microscopy. The combination of PDT and lipid nanoparticles has been evaluated in vitro regarding uptake, safety, and efficacy. The extensive and well-vascularized chorioallantois membrane (CAM) of fertilized hen's eggs offers an optimal platform for three-dimensional cell culture, which has been used in this study to evaluate the photodynamic efficacy of lipid nanoparticles against glioblastoma cells. In contrast to other animal models, the CAM model lacks a mature immune system in an early stage, facilitating the growth of xenografts without rejection. Treatment of xenografted U87 glioblastoma cells on CAM was performed to assess the effects on tumor viability, growth, and angiogenesis. The xenografts and the surrounding blood vessels were targeted through topical application, and the effects of photodynamic therapy have been confirmed microscopically and via positron emission tomography and X-ray computed tomography. Finally, the excised xenografts embedded in the CAM were analyzed histologically by hematoxylin and eosin and KI67 staining.
Collapse
Affiliation(s)
- Jan Schulze
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Lisa Schöne
- Institute
of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, Leipzig University, Eilenburger Strasse 15a, Leipzig 04317, Germany
| | - Abdallah M. Ayoub
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Damiano Librizzi
- Center
for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging,
Department of Nuclear Medicine, University
of Marburg, Hans-Meerwein-Strasse 3, Marburg 35043, Germany
| | - Muhammad Umair Amin
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Konrad Engelhardt
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Behrooz H. Yousefi
- Center
for Tumor Biology and Immunology (ZTI), Core Facility Molecular Imaging,
Department of Nuclear Medicine, University
of Marburg, Hans-Meerwein-Strasse 3, Marburg 35043, Germany
| | - Lena Bender
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Jens Schaefer
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Eduard Preis
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| | - Michaela Schulz-Siegmund
- Institute
of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, Leipzig University, Eilenburger Strasse 15a, Leipzig 04317, Germany
| | - Christian Wölk
- Institute
of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, Leipzig University, Eilenburger Strasse 15a, Leipzig 04317, Germany
| | - Udo Bakowsky
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Strasse 4, Marburg 35037, Germany
| |
Collapse
|
32
|
Li J, Brachtlova T, van der Meulen-Muileman IH, Kleerebezem S, Liu C, Li P, van Beusechem VW. Human Non-Small Cell Lung Cancer-Chicken Embryo Chorioallantoic Membrane Tumor Models for Experimental Cancer Treatments. Int J Mol Sci 2023; 24:15425. [PMID: 37895104 PMCID: PMC10607033 DOI: 10.3390/ijms242015425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
To promote the preclinical development of new treatments for non-small cell lung cancer (NSCLC), we established NSCLC xenograft tumor assays on the chorioallantoic membrane (CAM) of chicken embryos. Five NSCLC cell lines were compared for tumor take rate, tumor growth, and embryo survival. Two of these, A549 and H460 CAM tumors, were histologically characterized and tested for susceptibility to systemic chemotherapy and gene delivery using viral vectors. All cell lines were efficiently engrafted with minimal effect on embryo survival. The A549 cells formed slowly growing tumors, with a relatively uniform distribution of cancer cells and stroma cells, while the H460 cells formed large tumors containing mostly proliferating cancer cells in a bed of vascularized connective tissue. Tumor growth was inhibited via systemic treatment with Pemetrexed and Cisplatin, a chemotherapy combination that is often used to treat patients with advanced NSCLC. Lentiviral and adenoviral vectors expressing firefly luciferase transduced NSCLC tumors in vivo. The adenovirus vector yielded more than 100-fold higher luminescence intensities after a single administration than could be achieved with multiple lentiviral vector deliveries. The adenovirus vector also transduced CAM tissue and organs of developing embryos. Adenovirus delivery to tumors was 100-10,000-fold more efficient than to embryo organs. In conclusion, established human NSCLC-CAM tumor models provide convenient in vivo assays to rapidly evaluate new cancer therapies, particularly cancer gene therapies.
Collapse
Affiliation(s)
- Jing Li
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Tereza Brachtlova
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- ORCA Therapeutics BV, Onderwijsboulevard 225, 5223 DE 's Hertogenbosch, The Netherlands
| | - Ida H van der Meulen-Muileman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Stijn Kleerebezem
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Chang Liu
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Pulmonary Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Peiyu Li
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Victor W van Beusechem
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- ORCA Therapeutics BV, Onderwijsboulevard 225, 5223 DE 's Hertogenbosch, The Netherlands
| |
Collapse
|
33
|
de Campos WG, Araújo R, Júnior CAL, de Sousa Gomes P. Alendronate induces skeletal alterations in the chicken embryonic development model. Toxicol Appl Pharmacol 2023; 476:116673. [PMID: 37652309 DOI: 10.1016/j.taap.2023.116673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Alendronate, a nitrogen-containing bisphosphonate, has reported long-term clinical success in the management of distinct bone-related conditions, particularly in the modulation of post-menopausal osteoporosis. Nonetheless, whether the inhibitory activity over osteoclastic cells' functionality is widely acknowledged, contradictory evidence arises from the assessment of alendronate activity over osteoblastic populations. This may be of particular relevance in situations in which bone formation exceeds bone resorption, with further emphasis on embryonic development, since alendronate can cross the placental barrier and alendronate-based therapies are being extended into women of reproductive age. Accordingly, the present study aims to assess the effects of alendronate, at distinct concentrations (1.5E-10M to 1.5E-7M) on bone tissue development, within a translational animal model - the embryonic chicken development model. Embryos, at the beginning of osteogenesis (day 7) were exposed to different alendronate concentrations for 4 days. Embryos were following characterized for skeletal development by histomorphometric analysis upon histochemical staining, microtomographic analysis, and gene expression assessment of genes related to osteoclastogenic/osteoclastic and osteoblastogenic/osteogenic differentiation, as well as to the immuno-inflammatory activation. The findings revealed that exposure to alendronate had a dose-dependent impact on skeletal growth and mineralization. This effect was evidenced by diminished bone volume and reduced bone surface parameters, with the 1.5E-7M concentration leading to a remarkable reduction of over 50%. Additionally, a decreased osteoclastogenic/osteoclastic gene expression was verified, associated with a diminished osteoblastogenic/osteogenic program - within the 30-50% range for 1.5E-7 M, supporting the diminished bone formation process. An increased inflammatory activation may contribute, at least in part, to the attained outcomes. Overall present findings suggest a negative influence of alendronate on the embryonic bone development process in a dose-dependent manner, highlighting the potential risk of alendronate use during embryonic development.
Collapse
Affiliation(s)
| | - Rita Araújo
- Department of Stomatology, School of Dentistry, University of Sao Paulo, São Paulo, Brazil; Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
| | | | - Pedro de Sousa Gomes
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal.
| |
Collapse
|
34
|
Wessendorf J, Scheschenja M, Bastian MB, König AM, Pagenstecher A, Helmprobst F, Buchholz M, Tatura M, Jedelská J, Mahnken AH. Feasibility of the chick chorioallantoic membrane model for preclinical studies on tumor radiofrequency ablation. Eur Radiol Exp 2023; 7:56. [PMID: 37749303 PMCID: PMC10519884 DOI: 10.1186/s41747-023-00368-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/04/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND We evaluated the feasibility of a chick chorioallantoic membrane (CAM) tumor model for preclinical research on tumor radiofrequency ablation (RFA). METHODS Fertilized chicken eggs were incubated and divided into five cohorts: RFA for 30 s (n = 5), RFA for 60 s (n = 5), RFA for 120 s (n = 4), sham (n = 8), and controls (n = 6). Xenografting using pancreatic neuroendocrine tumor cells of the BON-1 cell line was performed on embryonic day (ED) 8. The RFA was performed on ED 12. Survival, stereomicroscopic observations, and histological observations using hematoxylin-eosin (H&E) and Ki67 staining were evaluated. RESULTS The survival rates in the 30-s, 60-s, and 120-s, sham and control cohort were 60%, 60%, 0%, 100%, and 50%, respectively. Signs of bleeding and heat damage were common findings in the evaluation of stereomicroscopic observations. Histological examination could be performed in all but one embryo. Heat damage, bleeding, thrombosis, and leukocyte infiltration and hyperemia were regular findings in H&E-stained cuts. A complete absence of Ki67 staining was recorded in 33.3% and 50% of embryos in the 30-s and 60-s cohorts that survived until ED 14, respectively. CONCLUSIONS The CAM model is a feasible and suiting research model for tumor RFA with many advantages over other animal models. It offers the opportunity to conduct in vivo research under standardized conditions. Further studies are needed to optimize this model for tumor ablations in order to explore promising but unrefined strategies like the combination of RFA and immunotherapy. RELEVANCE STATEMENT The chick chorioallantoic membrane model allows in vivo research on tumor radiofrequency ablation under standardized conditions that may enable enhanced understanding on combined therapies while ensuring animal welfare in concordance with the "Three Rs." KEY POINTS • The chorioallantoic membrane model is feasible and suiting for tumor radiofrequency ablation. • Radiofrequency ablation regularly achieved reduction but not eradication of Ki67 staining. • Histological evaluation showed findings comparable to changes in humans after RFA. • The chorioallantoic membrane model can enable studies on combined therapies after optimization.
Collapse
Affiliation(s)
- Joel Wessendorf
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany.
| | - Michael Scheschenja
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Moritz B Bastian
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Alexander M König
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Axel Pagenstecher
- Mouse Pathology and Electron Microscopy - Core Facility, Institute of Neuropathology, Philipps-University Marburg, Marburg, Germany
| | - Frederik Helmprobst
- Mouse Pathology and Electron Microscopy - Core Facility, Institute of Neuropathology, Philipps-University Marburg, Marburg, Germany
| | - Malte Buchholz
- Clinic for Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps-University Marburg, Marburg, Germany
| | - Marina Tatura
- Clinic for Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps-University Marburg, Marburg, Germany
| | - Jarmila Jedelská
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
- Small Animal MRI - Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Andreas H Mahnken
- Department of Diagnostic and Interventional Radiology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
- Small Animal MRI - Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
35
|
Reale A, Krutzke L, Cadamuro M, Vitiello A, von Einem J, Kochanek S, Palù G, Parolin C, Calistri A. Human Monocytes Are Suitable Carriers for the Delivery of Oncolytic Herpes Simplex Virus Type 1 In Vitro and in a Chicken Embryo Chorioallantoic Membrane Model of Cancer. Int J Mol Sci 2023; 24:ijms24119255. [PMID: 37298206 DOI: 10.3390/ijms24119255] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Oncolytic viruses (OVs) are promising therapeutics for tumors with a poor prognosis. An OV based on herpes simplex virus type 1 (oHSV-1), talimogene laherparepvec (T-VEC), has been recently approved by the Food and Drug Administration (FDA) and by the European Medicines Agency (EMA) for the treatment of unresectable melanoma. T-VEC, like most OVs, is administered via intratumoral injection, underlining the unresolved problem of the systemic delivery of the oncolytic agent for the treatment of metastases and deep-seated tumors. To address this drawback, cells with a tropism for tumors can be loaded ex vivo with OVs and used as carriers for systemic oncolytic virotherapy. Here, we evaluated human monocytes as carrier cells for a prototype oHSV-1 with a similar genetic backbone as T-VEC. Many tumors specifically recruit monocytes from the bloodstream, and autologous monocytes can be obtained from peripheral blood. We demonstrate here that oHSV-1-loaded primary human monocytes migrated in vitro towards epithelial cancer cells of different origin. Moreover, human monocytic leukemia cells selectively delivered oHSV-1 to human head-and-neck xenograft tumors grown on the chorioallantoic membrane (CAM) of fertilized chicken eggs after intravascular injection. Thus, our work shows that monocytes are promising carriers for the delivery of oHSV-1s in vivo, deserving further investigation in animal models.
Collapse
Affiliation(s)
- Alberto Reale
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Lea Krutzke
- Department of Gene Therapy, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Adriana Vitiello
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Jens von Einem
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| |
Collapse
|
36
|
Zou M, Wang T, Wang Y, Luo R, Sun Y, Peng X. Comparative Transcriptome Analysis Reveals the Innate Immune Response to Mycoplasma gallisepticum Infection in Chicken Embryos and Newly Hatched Chicks. Animals (Basel) 2023; 13:ani13101667. [PMID: 37238096 DOI: 10.3390/ani13101667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
Mycoplasma gallisepticum (MG) is a major cause of chronic respiratory diseases in chickens, with both horizontal and vertical transmission modes and varying degrees of impact on different ages. The innate immune response is crucial in resisting MG infection. Therefore, this study aimed to investigate the innate immune response of chicken embryos and newly hatched chicks to MG infection using comparative RNA-seq analysis. We found that MG infection caused weight loss and immune damage in both chicken embryos and chicks. Transcriptome sequencing analysis revealed that infected chicken embryos had a stronger immune response than chicks, as evidenced by the higher number of differentially expressed genes associated with innate immunity and inflammation. Toll-like receptor and cytokine-mediated pathways were the primary immune response pathways in both embryos and chicks. Furthermore, TLR7 signaling may play an essential role in the innate immune response to MG infection. Overall, this study sheds light on the development of innate immunity to MG infection in chickens and can help in devising disease control strategies.
Collapse
Affiliation(s)
- Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ronglong Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingfei Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
37
|
Jarrosson L, Dalle S, Costechareyre C, Tang Y, Grimont M, Plaschka M, Lacourrège M, Teinturier R, Le Bouar M, Maucort‐Boulch D, Eberhardt A, Castellani V, Caramel J, Delloye‐Bourgeois C. An in vivo avian model of human melanoma to perform rapid and robust preclinical studies. EMBO Mol Med 2023; 15:e16629. [PMID: 36692026 PMCID: PMC9994476 DOI: 10.15252/emmm.202216629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/25/2023] Open
Abstract
Metastatic melanoma patients carrying a BRAFV600 mutation can be treated with a combination of BRAF and MEK inhibitors (BRAFi/MEKi), but innate and acquired resistance invariably occurs. Predicting patient response to targeted therapies is crucial to guide clinical decision. We describe here the development of a highly efficient patient-derived xenograft model adapted to patient melanoma biopsies, using the avian embryo as a host (AVI-PDXTM ). In this in vivo paradigm, we depict a fast and reproducible tumor engraftment of patient samples within the embryonic skin, preserving key molecular and phenotypic features. We show that sensitivity and resistance to BRAFi/MEKi can be reliably modeled in these AVI-PDXTM , as well as synergies with other drugs. We further provide proof-of-concept that the AVI-PDXTM models the diversity of responses of melanoma patients to BRAFi/MEKi, within days, hence positioning it as a valuable tool for the design of personalized medicine assays and for the evaluation of novel combination strategies.
Collapse
Affiliation(s)
| | - Stéphane Dalle
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
- Centre Hospitalier Lyon SudHospices Civils de LyonPierre BéniteFrance
| | | | - Yaqi Tang
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
| | - Maxime Grimont
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
| | - Maud Plaschka
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
| | | | | | - Myrtille Le Bouar
- Centre Hospitalier Lyon SudHospices Civils de LyonPierre BéniteFrance
| | | | - Anaïs Eberhardt
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
- Centre Hospitalier Lyon SudHospices Civils de LyonPierre BéniteFrance
| | - Valérie Castellani
- University of Lyon, University of Lyon 1 Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene InstituteLyonFrance
| | - Julie Caramel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of LyonLyonFrance
| | - Céline Delloye‐Bourgeois
- University of Lyon, University of Lyon 1 Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene InstituteLyonFrance
| |
Collapse
|
38
|
Ribatti D. The chick embryo chorioallantoic membrane patient-derived xenograft (PDX) model. Pathol Res Pract 2023; 243:154367. [PMID: 36774760 DOI: 10.1016/j.prp.2023.154367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
The chick embryo chorioallantoic membrane (CAM) CAM is an extraembryonic membrane generated by the fusion of the chorion with the vascularized allantoic membrane. It performs multiple functions during embryonic development, including respiration, calcium transport from the eggshell, acid-base homeostasis, and ion/water reabsorption from the allantoic fluid. The CAM is a widely used model for the study of angiogenesis, anti-angiogenesis, tumor growth, and metastasis as well as drug efficacy. Ethical approval is omitted if experiments are terminated at embryonic day 14 in most countries, facilitating screenings of pharmacological or physics-based therapies with high reproducibility at large scales supporting the 3Rs principle. Being naturally immunodeficient, the chick embryo accepts transplantation from various tissues and species without immune response. This review article is focused on the analysis of the literature and personal data concerning the effects of patient-derived xenografts (PDX) on the CAM.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
39
|
Oliinyk D, Eigenberger A, Felthaus O, Haerteis S, Prantl L. Chorioallantoic Membrane Assay at the Cross-Roads of Adipose-Tissue-Derived Stem Cell Research. Cells 2023; 12:cells12040592. [PMID: 36831259 PMCID: PMC9953848 DOI: 10.3390/cells12040592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
With a history of more than 100 years of different applications in various scientific fields, the chicken chorioallantoic membrane (CAM) assay has proven itself to be an exceptional scientific model that meets the requirements of the replacement, reduction, and refinement principle (3R principle). As one of three extraembryonic avian membranes, the CAM is responsible for fetal respiration, metabolism, and protection. The model provides a unique constellation of immunological, vascular, and extracellular properties while being affordable and reliable at the same time. It can be utilized for research purposes in cancer biology, angiogenesis, virology, and toxicology and has recently been used for biochemistry, pharmaceutical research, and stem cell biology. Stem cells and, in particular, mesenchymal stem cells derived from adipose tissue (ADSCs) are emerging subjects for novel therapeutic strategies in the fields of tissue regeneration and personalized medicine. Because of their easy accessibility, differentiation profile, immunomodulatory properties, and cytokine repertoire, ADSCs have already been established for different preclinical applications in the files mentioned above. In this review, we aim to highlight and identify some of the cross-sections for the potential utilization of the CAM model for ADSC studies with a focus on wound healing and tissue engineering, as well as oncological research, e.g., sarcomas. Hereby, the focus lies on the combination of existing evidence and experience of such intersections with a potential utilization of the CAM model for further research on ADSCs.
Collapse
Affiliation(s)
- Dmytro Oliinyk
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
- Correspondence:
| | - Andreas Eigenberger
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, Faculty for Biology and Preclinical Medicine, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
40
|
Sun X, Wang Y, Wang C, Wang Y, Ren Z, Yang X, Yang X, Liu Y. Genome analysis reveals hepatic transcriptional reprogramming changes mediated by enhancers during chick embryonic development. Poult Sci 2023; 102:102516. [PMID: 36764138 PMCID: PMC9929590 DOI: 10.1016/j.psj.2023.102516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The liver undergoes a slow process for lipid deposition during chick embryonic period. However, the underlying physiological and molecular mechanisms are still unclear. Therefore, the aim of the current study was to reveal the epigenetic mechanism of hepatic transcriptional reprogramming changes based on the integration analysis of RNA-seq and H3K27ac labeled CUT&Tag. Results showed that lipid contents increased gradually with the embryonic age (E) 11, E15, and E19 based on morphological analysis of Hematoxylin-eosin and Oil Red O staining as well as total triglyceride and cholesterol detection. The hepatic protein level of SREBP-1c was higher in E19 when compared with that in E11 and E15, while H3K27ac and H3K4me2 levels declined from E11 to E19. Differential expression genes (DEGs) among these 3 embryonic ages were determined by transcriptome analysis. A total of 107 and 46 genes were gradually upregulated and downregulated respectively with the embryonic age. Meanwhile, differential H3K27ac occupancy in chromatin was investigated. But the integration analysis of RNA-seq and CUT&Tag data showed that the overlap genes were less between DEGs and target genes of differential peaks in the promoter regions. Further, some KEGG pathways enriched from target genes of typical enhancer were overlapped with those from DEGs in transcriptome analysis such as insulin, FoxO, MAPK signaling pathways which were related to lipid metabolism. DNA motif analysis identify 8 and 10 transcription factors (TFs) based on up and down differential peaks individually among E11, E15, and E19 stages where 7 TFs were overlapped including COUP-TFII, FOXM1, FOXA1, HNF4A, RXR, ERRA, FOXA2. These results indicated that H3K27ac histone modification is involved in the transcriptional reprogramming regulation during embryonic development, which could recruit TFs binding to mediate differential enhancer activation. Differential activated enhancer impels dynamic transcriptional reprogramming towards lipid metabolism to promote the occurrence of special phenotype of hepatic lipid deposition.
Collapse
Affiliation(s)
- Xi Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yumeng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chaohui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhouzheng Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
41
|
Negi V, Kuhn RJ, Fekete DM. Exploring the Expression and Function of cTyro3, a Candidate Zika Virus Receptor, in the Embryonic Chicken Brain and Inner Ear. Viruses 2023; 15:247. [PMID: 36680287 PMCID: PMC9867072 DOI: 10.3390/v15010247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The transmembrane protein Axl was proposed as an entry receptor for Zika virus (ZIKV) infection in vitro, but conflicting results from in vivo studies have made it difficult to establish Axl as a physiologically relevant ZIKV receptor. Both the functional redundancy of receptors and the experimental model used can lead to variable results. Therefore, it can be informative to explore alternative animal models to analyze ZIKV receptor candidates as an aid in discovering antivirals. This study used chicken embryos to examine the role of chicken Tyro3 (cTyro3), the equivalent of human Axl. Results show that endogenous cTyro3 mRNA expression overlaps with previously described hot spots of ZIKV infectivity in the brain and inner ear. We asked if ectopic expression or knockdown of cTyro3 influenced ZIKV infection in embryos. Tol2 vectors or replication-competent avian retroviruses were used in ovo to introduce full-length or truncated (presumed dominant-negative) cTyro3, respectively, into the neural tube on embryonic day two (E2). ZIKV was delivered to the brain 24 h later. cTyro3 manipulations did not alter ZIKV infection or cell death in the E5/E6 brain. Moreover, delivery of truncated cTyro3 variants to the E3 otocyst had no effect on inner ear formation on E6 or E10.
Collapse
Affiliation(s)
| | | | - Donna M. Fekete
- Department of Biological Sciences, Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
42
|
Fischer D, Fluegen G, Garcia P, Ghaffari-Tabrizi-Wizsy N, Gribaldo L, Huang RYJ, Rasche V, Ribatti D, Rousset X, Pinto MT, Viallet J, Wang Y, Schneider-Stock R. The CAM Model-Q&A with Experts. Cancers (Basel) 2022; 15:cancers15010191. [PMID: 36612187 PMCID: PMC9818221 DOI: 10.3390/cancers15010191] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
The chick chorioallantoic membrane (CAM), as an extraembryonic tissue layer generated by the fusion of the chorion with the vascularized allantoic membrane, is easily accessible for manipulation. Indeed, grafting tumor cells on the CAM lets xenografts/ovografts develop in a few days for further investigations. Thus, the CAM model represents an alternative test system that is a simple, fast, and low-cost tool to study tumor growth, drug response, or angiogenesis in vivo. Recently, a new era for the CAM model in immune-oncology-based drug discovery has been opened up. Although there are many advantages offering extraordinary and unique applications in cancer research, it has also disadvantages and limitations. This review will discuss the pros and cons with experts in the field.
Collapse
Affiliation(s)
- Dagmar Fischer
- Division of Pharmaceutical Technology, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Georg Fluegen
- Department of General, Visceral, Thoracic and Pediatric Surgery (A), Medical Faculty, Heinrich-Heine-University, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Paul Garcia
- Institute for Advanced Biosciences, Research Center Université Grenoble Alpes (UGA)/Inserm U 1209/CNRS 5309, 38700 La Tronche, France
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Nassim Ghaffari-Tabrizi-Wizsy
- SFL Chicken CAM Lab, Department of Immunology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Laura Gribaldo
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Volker Rasche
- Department of Internal Medicine II, Ulm University Medical Center, 89073 Ulm, Germany
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neurosciences, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Marta Texeira Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Ipatimup—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - Jean Viallet
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Yan Wang
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 94054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8526-069
| |
Collapse
|
43
|
Butler K, Brinker CJ, Leong HS. Bridging the In Vitro to In Vivo gap: Using the Chick Embryo Model to Accelerate Nanoparticle Validation and Qualification for In Vivo studies. ACS NANO 2022; 16:19626-19650. [PMID: 36453753 PMCID: PMC9799072 DOI: 10.1021/acsnano.2c03990] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
We postulate that nanoparticles (NPs) for use in therapeutic applications have largely not realized their clinical potential due to an overall inability to use in vitro results to predict NP performance in vivo. The avian embryo and associated chorioallantoic membrane (CAM) has emerged as an in vivo preclinical model that bridges the gap between in vitro and in vivo, enabling rapid screening of NP behavior under physiologically relevant conditions and providing a rapid, accessible, economical, and more ethical means of qualifying nanoparticles for in vivo use. The CAM is highly vascularized and mimics the diverging/converging vasculature of the liver, spleen, and lungs that serve as nanoparticle traps. Intravital imaging of fluorescently labeled NPs injected into the CAM vasculature enables immediate assessment and quantification of nano-bio interactions at the individual NP scale in any tissue of interest that is perfused with a microvasculature. In this review, we highlight how utilization of the avian embryo and its CAM as a preclinical model can be used to understand NP stability in blood and tissues, extravasation, biocompatibility, and NP distribution over time, thereby serving to identify a subset of NPs with the requisite stability and performance to introduce into rodent models and enabling the development of structure-property relationships and NP optimization without the sacrifice of large populations of mice or other rodents. We then review how the chicken embryo and CAM model systems have been used to accelerate the development of NP delivery and imaging agents by allowing direct visualization of targeted (active) and nontargeted (passive) NP binding, internalization, and cargo delivery to individual cells (of relevance for the treatment of leukemia and metastatic cancer) and cellular ensembles (e.g., cancer xenografts of interest for treatment or imaging of cancer tumors). We conclude by showcasing emerging techniques for the utilization of the CAM in future nano-bio studies.
Collapse
Affiliation(s)
- Kimberly
S. Butler
- Molecular
and Microbiology, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - C. Jeffrey Brinker
- Department
of Chemical and Biological Engineering and the Comprehensive Cancer
Center, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Hon Sing Leong
- Department
of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto M5G 1L7, Canada
- Biological
Sciences Platform, Sunnybrook Hospital, Toronto M4N 3M5, Canada
| |
Collapse
|
44
|
Devaux CA, Pontarotti P, Nehari S, Raoult D. 'Cannibalism' of exogenous DNA sequences: The ancestral form of adaptive immunity which entails recognition of danger. Front Immunol 2022; 13:989707. [PMID: 36618387 PMCID: PMC9816338 DOI: 10.3389/fimmu.2022.989707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Adaptive immunity is a sophisticated form of immune response capable of retaining the molecular memory of a very great diversity of target antigens (epitopes) as non-self. It is capable of reactivating itself upon a second encounter with an immunoglobulin or T-cell receptor antigen-binding site with a known epitope that had previously primed the host immune system. It has long been considered that adaptive immunity is a highly evolved form of non-self recognition that appeared quite late in speciation and complemented a more generalist response called innate immunity. Innate immunity offers a relatively non-specific defense (although mediated by sensors that could specifically recognize virus or bacteria compounds) and which does not retain a memory of the danger. But this notion of recent acquisition of adaptive immunity is challenged by the fact that another form of specific recognition mechanisms already existed in prokaryotes that may be able to specifically auto-protect against external danger. This recognition mechanism can be considered a primitive form of specific (adaptive) non-self recognition. It is based on the fact that many archaea and bacteria use a genome editing system that confers the ability to appropriate viral DNA sequences allowing prokaryotes to prevent host damage through a mechanism very similar to adaptive immunity. This is indistinctly called, 'endogenization of foreign DNA' or 'viral DNA predation' or, more pictorially 'DNA cannibalism'. For several years evidence has been accumulating, highlighting the crucial role of endogenization of foreign DNA in the fundamental processes related to adaptive immunity and leading to a change in the dogma that adaptive immunity appeared late in speciation.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille University, Institut de recherche pour le développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), MEPHI, Institut Hospitalo-universitaire (IHU)-Méditerranée Infection, Marseille, France,Department of Biological Sciences, Centre National de la Recherche Scientifique, Centre National de la Recherche Scientifique (CNRS)-SNC5039, Marseille, France,*Correspondence: Christian A. Devaux,
| | - Pierre Pontarotti
- Aix-Marseille University, Institut de recherche pour le développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), MEPHI, Institut Hospitalo-universitaire (IHU)-Méditerranée Infection, Marseille, France,Department of Biological Sciences, Centre National de la Recherche Scientifique, Centre National de la Recherche Scientifique (CNRS)-SNC5039, Marseille, France
| | - Sephora Nehari
- Aix-Marseille University, Institut de recherche pour le développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), MEPHI, Institut Hospitalo-universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille University, Institut de recherche pour le développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), MEPHI, Institut Hospitalo-universitaire (IHU)-Méditerranée Infection, Marseille, France
| |
Collapse
|
45
|
Barnett SE, Herrmann A, Shaw L, Gash EN, Poptani H, Sacco JJ, Coulson JM. The Chick Embryo Xenograft Model for Malignant Pleural Mesothelioma: A Cost and Time Efficient 3Rs Model for Drug Target Evaluation. Cancers (Basel) 2022; 14:5836. [PMID: 36497318 PMCID: PMC9740959 DOI: 10.3390/cancers14235836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) has limited treatment options and poor prognosis. Frequent inactivation of the tumour suppressors BAP1, NF2 and P16 may differentially sensitise tumours to treatments. We have established chick chorioallantoic membrane (CAM) xenograft models of low-passage MPM cell lines and protocols for evaluating drug responses. Ten cell lines, representing the spectrum of histological subtypes and tumour suppressor status, were dual labelled for fluorescence/bioluminescence imaging and implanted on the CAM at E7. Bioluminescence was used to assess viability of primary tumours, which were excised at E14 for immunohistological staining or real-time PCR. All MPM cell lines engrafted efficiently forming vascularised nodules, however their size, morphology and interaction with chick cells varied. MPM phenotypes including local invasion, fibroblast recruitment, tumour angiogenesis and vascular remodelling were evident. Bioluminescence imaging could be used to reliably estimate tumour burden pre- and post-treatment, correlating with tumour weight and Ki-67 staining. In conclusion, MPM-CAM models recapitulate important features of the disease and are suitable to assess drug targets using a broad range of MPM cell lines that allow histological or genetic stratification. They are amenable to multi-modal imaging, potentially offering a time and cost-efficient, 3Rs-compliant alternative to rodent xenograft models to prioritise candidate compounds from in vitro studies.
Collapse
Affiliation(s)
- Sarah E. Barnett
- Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
- Technology, Infrastructure & Environment Directorate, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L69 7ZX, UK
| | - Anne Herrmann
- Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Liam Shaw
- Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Elisabeth N. Gash
- Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Harish Poptani
- Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Joseph J. Sacco
- Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Judy M. Coulson
- Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
46
|
Integrated Transcriptome Analysis Reveals mRNA-miRNA Pathway Crosstalk in Roman Laying Hens' Immune Organs Induced by AFB1. Toxins (Basel) 2022; 14:toxins14110808. [PMID: 36422982 PMCID: PMC9693605 DOI: 10.3390/toxins14110808] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a widely distributed contaminant in moldy corn, rice, soybean, and oil crops. Many studies have revealed its adverse effects, such as carcinogenicity, immunotoxicity, and hepatotoxicity, on the health of humans and animals. To investigate the immunotoxic effects on chicken immune organs induced by AFB1, we integrated RNA and small-RNA sequencing data of the spleen and the bursa of Fabricius to elucidate the response of the differentially expressed transcriptional profiles and related pathways. AFB1 consumption negatively influenced egg quality, but no obvious organ damage was observed compared to that of the control group. We identified 3918 upregulated and 2415 downregulated genes in the spleen and 231 upregulated and 65 downregulated genes in the bursa of Fabricius. We confirmed that several core genes related to immune and metabolic pathways were activated by AFB1. Furthermore, 42 and 19 differentially expressed miRNAs were found in the spleen and the bursa of Fabricius, respectively. Differentially expressed genes and target genes of differentially expressed miRNAs were mainly associated with cancer progression and immune response. The predicted mRNA-miRNA pathway network illustrated the potential regulatory mechanisms. The present study identified the transcriptional profiles and revealed potential mRNA-miRNA pathway crosstalk. This genetic regulatory network will facilitate the understanding of the immunotoxicity mechanisms of chicken immune organs induced by high concentrations of AFB1.
Collapse
|
47
|
Santos JC, Profitós-Pelejà N, Ribeiro ML, Roué G. Antitumor Activity of Simvastatin in Preclinical Models of Mantle Cell Lymphoma. Cancers (Basel) 2022; 14:cancers14225601. [PMID: 36428695 PMCID: PMC9688202 DOI: 10.3390/cancers14225601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/02/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a rare and aggressive subtype of B-cell non-Hodgkin lymphoma that remains incurable with standard therapy. Statins are well-tolerated, inexpensive, and widely prescribed as cholesterol-lowering agents to treat hyperlipidemia and to prevent cardiovascular diseases through the blockage of the mevalonate metabolic pathway. These drugs have also shown promising anti-cancer activity through pleiotropic effects including the induction of lymphoma cell death. However, their potential use as anti-MCL agents has not been evaluated so far. AIM The present study aimed to investigate the activity of simvastatin on MCL cells. METHODS We evaluated the cytotoxicity of simvastatin in MCL cell lines by CellTiter-Glo and lactate dehydrogenase (LDH) release assays. Cell proliferation and mitotic index were assessed by direct cell recounting and histone H3-pSer10 immunostaining. Apoptosis induction and reactive oxygen species (ROS) generation were evaluated by flow cytometry. Cell migration and invasion properties were determined by transwell assay. The antitumoral effect of simvastatin in vivo was evaluated in a chick embryo chorioallantoic membrane (CAM) MCL xenograft model. RESULTS We show that treatment with simvastatin induced a 2 to 6-fold LDH release, inhibited more than 50% of cell proliferation, and enhanced the caspase-independent ROS-mediated death of MCL cells. The effective impairment of MCL cell survival was accompanied by the inhibition of AKT and mTOR phosphorylation. Moreover, simvastatin strongly decreased MCL cell migration and invasion ability, leading to a 55% tumor growth inhibition and a consistent diminution of bone marrow and spleen metastasis in vivo. CONCLUSION Altogether, these data provide the first preclinical insight into the effect of simvastatin against MCL cells, suggesting that this agent might be considered for repurpose as a precise MCL therapy.
Collapse
Affiliation(s)
- Juliana Carvalho Santos
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
- Correspondence: (J.C.S.); (G.R.); Tel.: +34-935572800 (ext. 4081) (J.C.S.); +34-935572835 (G.R.)
| | - Núria Profitós-Pelejà
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
| | - Marcelo Lima Ribeiro
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista 01246-100, SP, Brazil
| | - Gaël Roué
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, 08916 Badalona, Spain
- Correspondence: (J.C.S.); (G.R.); Tel.: +34-935572800 (ext. 4081) (J.C.S.); +34-935572835 (G.R.)
| |
Collapse
|
48
|
Sola B, Caillot M. L’embryon de poule. Med Sci (Paris) 2022; 38:795-799. [DOI: 10.1051/medsci/2022123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Le développement de drogues anti-cancéreuses à visée thérapeutique nécessite leur évaluation. Ces drogues candidates sont généralement testées in vitro, sur des lignées cellulaires ou sur des cellules isolées à partir de patients, et, in vivo, dans des modèles de xénogreffe chez la souris immunodéprimée. Depuis quelques années, les contraintes réglementaires (règle des 3R : réduire, raffiner, remplacer) imposent de mettre en place des modèles alternatifs qui se substituent aux modèles murins ou, au moins, en limitent l’utilisation. Parmi les modèles alternatifs proposés, la greffe sur membrane chorio-allantoïdienne d’embryon de poule semble performante. Elle permet de suivre et de quantifier la croissance tumorale et d’autres paramètres associés, comme la néo-angiogenèse, l’invasion et la migration tumorales. Elle permet aussi le criblage de drogues. Ce modèle semble également adapté à la médecine personnalisée en cancérologie. Nous présentons dans cette revue la technique et ses avantages.
Collapse
|
49
|
Miebach L, Berner J, Bekeschus S. In ovo model in cancer research and tumor immunology. Front Immunol 2022; 13:1006064. [PMID: 36248802 PMCID: PMC9556724 DOI: 10.3389/fimmu.2022.1006064] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Considering cancer not only as malignant cells on their own but as a complex disease in which tumor cells interact and communicate with their microenvironment has motivated the establishment of clinically relevant 3D models in past years. Technological advances gave rise to novel bioengineered models, improved organoid systems, and microfabrication approaches, increasing scientific importance in preclinical research. Notwithstanding, mammalian in vivo models remain closest to mimic the patient’s situation but are limited by cost, time, and ethical constraints. Herein, the in ovo model bridges the gap as an advanced model for basic and translational cancer research without the need for ethical approval. With the avian embryo being a naturally immunodeficient host, tumor cells and primary tissues can be engrafted on the vascularized chorioallantoic membrane (CAM) with high efficiencies regardless of species-specific restrictions. The extraembryonic membranes are connected to the embryo through a continuous circulatory system, readily accessible for manipulation or longitudinal monitoring of tumor growth, metastasis, angiogenesis, and matrix remodeling. However, its applicability in immunoncological research is largely underexplored. Dual engrafting of malignant and immune cells could provide a platform to study tumor-immune cell interactions in a complex, heterogenic and dynamic microenvironment with high reproducibility. With some caveats to keep in mind, versatile methods for in and ex ovo monitoring of cellular and molecular dynamics already established in ovo are applicable alike. In this view, the present review aims to emphasize and discuss opportunities and limitations of the chicken embryo model for pre-clinical research in cancer and cancer immunology.
Collapse
Affiliation(s)
- Lea Miebach
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
| | - Julia Berner
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, Greifswald University Medical Center, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
- *Correspondence: Sander Bekeschus,
| |
Collapse
|
50
|
The Comparative Experimental Study of Sodium and Magnesium Dichloroacetate Effects on Pediatric PBT24 and SF8628 Cell Glioblastoma Tumors Using a Chicken Embryo Chorioallantoic Membrane Model and on Cells In Vitro. Int J Mol Sci 2022; 23:ijms231810455. [PMID: 36142368 PMCID: PMC9499689 DOI: 10.3390/ijms231810455] [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: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, pyruvate dehydrogenase kinase-1 inhibition with dichloroacetate (DCA) was explored as an alternative cancer therapy. The study’s aim was to compare the effectiveness of NaDCA and MgDCA on pediatric glioblastoma PBT24 and SF8628 tumors and cells. The treatment effects were evaluated on xenografts growth on a chicken embryo chorioallantoic membrane. The PCNA, EZH2, p53, survivin expression in tumor, and the SLC12A2, SLC12A5, SLC5A8, CDH1, and CDH2 expression in cells were studied. The tumor groups were: control, cells treated with 10 mM and 5 mM of NaDCA, and 5 mM and 2.5 mM of MgDCA. The cells were also treated with 3 mM DCA. Both the 10 mM DCA preparations significantly reduced PBT24 and SF8624 tumor invasion rates, while 5 mM NaDCA reduced it only in the SF8628 tumors. The 5 mM MgDCA inhibited tumor-associated neoangiogenesis in PBT24; both doses of NaDCA inhibited tumor-associated neoangiogenesis in SF8628. The 10 mM DCA inhibited the expression of markers tested in PBT24 and SF8628 tumors, but the 5 mM DCA affect on their expression depended on the cation. The DCA treatment did not affect the SLC12A2, SLC12A5, and SLC5A8 expression in cells but increased CDH1 expression in SF8628. The tumor response to DCA at different doses indicated that a contrast between NaDCA and MgDCA effectiveness reflects the differences in the tested cells’ biologies.
Collapse
|