1
|
Oh JH, Karadeniz F, Seo Y, Kong CS. Isopimpinellin inhibits UVA-induced overproduction of MMPs via suppression of MAPK/AP-1 signaling in human dermal fibroblasts. Food Sci Biotechnol 2024; 33:3579-3589. [PMID: 39493386 PMCID: PMC11525369 DOI: 10.1007/s10068-024-01611-2] [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: 01/16/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 11/05/2024] Open
Abstract
Corydalis heterocarpa is an edible halophyte and an ingredient in traditional Korean medicine. In the present study, isopimpinellin (IPN), a bioactive coumarin, was isolated from the medicinal halophyte C. heterocarpa, and the effects of IPN against UVA-induced photoaging were investigated in human dermal fibroblasts. Photoaging is a skin disorder that manifests itself as premature skin aging due to chronic exposure to UV radiation. The symptoms of photoaging mainly arise from degraded skin connective tissue produced by overly expressed matrix metalloproteinases (MMPs). IPN treatment decreased the UVA-induced formation of reactive oxygen species and decreased MMP-1, MMP-3, and MMP-9 collagenases at the protein level. The UVA-mediated suppression of tissue inhibitors of MMP-1 and -2 was attenuated with IPN. The presence of 10 μM IPN inhibited the MAPK-mediated phosphorylation of c-Fos and c-Jun. In conclusion, the overall result of the current study indicated that IPN inhibited the UVA-induced overexpression of MMPs via blocking the MAPK/AP-1 pathway.
Collapse
Affiliation(s)
- Jung Hwan Oh
- Marine Biotechnology Center for Pharmaceuticals and Foods, College of Medical and Life Sciences, Silla University, Busan, 46958 Republic of Korea
- Nutritional Education, Graduate School of Education, Silla University, Busan, 46958 Republic of Korea
| | - Fatih Karadeniz
- Marine Biotechnology Center for Pharmaceuticals and Foods, College of Medical and Life Sciences, Silla University, Busan, 46958 Republic of Korea
| | - Youngwan Seo
- Division of Convergence on Marine Science, College of Ocean Science and Technology, Korea Maritime and Ocean University, Busan, 49112 Republic of Korea
| | - Chang-Suk Kong
- Marine Biotechnology Center for Pharmaceuticals and Foods, College of Medical and Life Sciences, Silla University, Busan, 46958 Republic of Korea
- Department of Food and Nutrition, College of Medical and Life Sciences, Silla University, Busan, 46958 Republic of Korea
| |
Collapse
|
2
|
Bartnik M, Sławińska-Brych A, Mizerska-Kowalska M, Kania AK, Zdzisińska B. Quantitative Analysis of Isopimpinellin from Ammi majus L. Fruits and Evaluation of Its Biological Effect on Selected Human Tumor Cells. Molecules 2024; 29:2874. [PMID: 38930940 PMCID: PMC11206288 DOI: 10.3390/molecules29122874] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Ammi majus L. (Apiaceae) is a medicinal plant with a well-documented history in phytotherapy. The aim of the present work was to isolate isopimpinellin (5,8-methoxypsoralen; IsoP) from the fruit of this plant and evaluate its biological activity against selected tumor cell lines. The methanol extract obtained with the use of an accelerated solvent extraction (ASE) method was the most suitable for the quantitative analysis of coumarins in the A. majus fruit matrix. The coumarin content was estimated by RP-HPLC/DAD, and the amount of IsoP was found to be 404.14 mg/100 g dry wt., constituting 24.56% of the total coumarin fraction (1.65 g/100 g). This, along with the presence of xanthotoxin (368.04 mg/100 g, 22.36%) and bergapten (253.05 mg/100 g, 15.38%), confirmed A. majus fruits as an excellent source of these compounds. IsoP was isolated (99.8% purity) by combined liquid chromatography/centrifugal partition chromatography (LC/CPC) and tested for the first time on its antiproliferative activity against human colorectal adenocarcinoma (HT29, SW620), osteosarcoma (Saos-2, HOS), and multiple myeloma (RPMI8226, U266) cell lines. MTT assay results (96 h incubation) demonstrated a dose- and cell line-dependent decrease in cell proliferation/viability, with the strongest effect of IsoP against the Saos-2 cell line (IC50; 42.59 µM), medium effect against U266, HT-29, and RPMI8226 (IC50 = 84.14, 95.53, and 105.0 µM, respectively), and very weak activity against invasive HOS (IC50; 321.6 µM) and SW620 (IC50; 711.30 µM) cells, as well as normal human skin fibroblasts (HSFs), with IC50; 410.7 µM. The mechanistic study on the Saos-2 cell line showed that IsoP was able to reduce DNA synthesis and trigger apoptosis via caspase-3 activation. In general, IsoP was found to have more potency towards cancerous cells (except for HOS and SW620) than against healthy cells. The Selective Index (SI) was determined, underlining the higher selectivity of IsoP towards cancer cells compared to healthy cells (SI = 9.62 against Saos-2). All these results suggest that IsoP might be a promising molecule in the chemo-prevention and treatment of primary osteosarcoma.
Collapse
Affiliation(s)
- Magdalena Bartnik
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodźki 1 Street, 20-093 Lublin, Poland;
| | - Adrianna Sławińska-Brych
- Department of Cell Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland;
| | - Magdalena Mizerska-Kowalska
- Department of Virology and Immunology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (M.M.-K.); (B.Z.)
| | - Anna Karolina Kania
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodźki 1 Street, 20-093 Lublin, Poland;
| | - Barbara Zdzisińska
- Department of Virology and Immunology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (M.M.-K.); (B.Z.)
| |
Collapse
|
3
|
Liu L, Cai F, Lu Y, Xie Y, Li H, Long C. Comparative Lipidomic and Metabolomic Analyses Reveal the Mystery of Lacquer Oil from Toxicodendron vernicifluum for the Treatment of “Yuezi” Disease in Nujiang, China: From Anti-Inflammation and Anti-Postpartum Depression Perspective. Front Pharmacol 2022; 13:914951. [PMID: 35770099 PMCID: PMC9234167 DOI: 10.3389/fphar.2022.914951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In southwest China, especially in Nujiang, lacquer oil from the drupes of Toxicodendron vernicifluum (Stokes) F. A. Barkley, including black lacquer oil (BLO) and white lacquer oil (WLO), is one of the most important edible oils for the local people. Through the field investigation, the locals believe that lacquer oil has benefits for parturient women and for the treatment of “Yuezi” disease. However, studies on bioactivities and the chemical compositions of lacquer oil are limited.Purpose: This study was designed to reveal the mystery of lacquer oil for the treatment of “Yuezi” disease by testing its anti-inflammatory and anti-postpartum depressant activities and related bioactive compounds.Methods: The anti-inflammatory effects of lacquer oil were examined by establishing a lipopolysaccharide (LPS)-induced RAW264.7 cell inflammation model and detecting the level of pro-inflammatory factors such as NO, IL-6 and TNF-α. The antidepressant effects of lacquer oil were studied by building a mouse model of postpartum depression (PPD), and the animal behavior changes of PPD model mice were assessed by open field test (OFT), forced swimming test (FST) and tail suspension test (TST). The chemical profiles of BLO and WLO were detected by lipidomic and the untargeted metabolomic research methods based on UPLC-MS/MS.Results: The results showed that BLO and WLO exerted anti-inflammatory effects by reducing the release of pro-inflammatory factors and BLO had better anti-inflammatory effects than WLO. While only BLO had anti-postpartum depressant activities, as evidenced by the significantly reduced the immobility time of the BLO-treated PPD mice in TST and FST compared to the PPD model mice. The comparative lipidomic analysis revealed that BLO contained high levels of Diacylglycerols (DAG) and Diacylglyceryl trimethylhomoserines (DGTS) but low level of ceramides (Cer), sphingomyelines (SM), phosphatidylcholines (PC) and phosphatidylethanolamines (PE) compared with WLO. Metabolomics analysis showed that there were 57 chemical markers between BLO and WLO, of which 17 potential biomarkers have been declared to possess anti-inflammatory and/or antidepressant activities.Conclusion: The findings of this study furnish a scientific support for the traditional uses of lacquer oil for the treatment of “Yuezi” disease from anti-inflammation and anti-postpartum depression perspective.
Collapse
Affiliation(s)
- Liya Liu
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Fei Cai
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Yitong Lu
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yuting Xie
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Hao Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Chunlin Long
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
- *Correspondence: Chunlin Long, ,
| |
Collapse
|
4
|
Geurtzen K, López-Delgado AC, Duseja A, Kurzyukova A, Knopf F. Laser-mediated osteoblast ablation triggers a pro-osteogenic inflammatory response regulated by reactive oxygen species and glucocorticoid signaling in zebrafish. Development 2022; 149:275194. [DOI: 10.1242/dev.199803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022]
Abstract
ABSTRACT
In zebrafish, transgenic labeling approaches, robust regenerative responses and excellent in vivo imaging conditions enable precise characterization of immune cell behavior in response to injury. Here, we monitored osteoblast-immune cell interactions in bone, a tissue which is particularly difficult to in vivo image in tetrapod species. Ablation of individual osteoblasts leads to recruitment of neutrophils and macrophages in varying numbers, depending on the extent of the initial insult, and initiates generation of cathepsin K+ osteoclasts from macrophages. Osteoblast ablation triggers the production of pro-inflammatory cytokines and reactive oxygen species, which are needed for successful macrophage recruitment. Excess glucocorticoid signaling as it occurs during the stress response inhibits macrophage recruitment, maximum speed and changes the macrophage phenotype. Although osteoblast loss is compensated for within a day by contribution of committed osteoblasts, macrophages continue to populate the region. Their presence is required for osteoblasts to fill the lesion site. Our model enables visualization of bone repair after microlesions at single-cell resolution and demonstrates a pro-osteogenic function of tissue-resident macrophages in non-mammalian vertebrates.
Collapse
Affiliation(s)
- Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ankita Duseja
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Department of Oncology and Metabolism, Metabolic Bone Centre, Sorby Wing, Northern General Hospital, Sheffield S5 7AU, UK
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Faculty of Health and Medical Sciences, Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
5
|
Design and Synthesis of Arylpiperazine Serotonergic/Dopaminergic Ligands with Neuroprotective Properties. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041297. [PMID: 35209087 PMCID: PMC8877291 DOI: 10.3390/molecules27041297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/21/2022]
Abstract
Long-chain arylpiperazine scaffold is a versatile template to design central nervous system (CNS) drugs that target serotonin and dopamine receptors. Here we describe the synthesis and biological evaluation of ten new arylpiperazine derivatives designed to obtain an affinity profile at serotonin 5-HT1A, 5-HT2A, 5-HT7 receptor, and dopamine D2 receptor of prospective drugs to treat the core symptoms of autism spectrum disorder (ASD) or psychosis. Besides the structural features required for affinity at the target receptors, the new compounds incorporated structural fragments with antioxidant properties to counteract oxidative stress connected with ASD and psychosis. All the new compounds showed CNS MultiParameter Optimization score predictive of desirable ADMET properties and cross the blood–brain barrier. We identified compound 12a that combines an affinity profile compatible with antipsychotic activity (5-HT1AKi = 41.5 nM, 5-HT2AKi = 315 nM, 5-HT7Ki = 42.5 nM, D2Ki = 300 nM), and compound 9b that has an affinity profile consistent with studies in the context of ASD (5-HT1AKi = 23.9 nM, 5-HT2AKi = 39.4 nM, 5-HT7Ki = 45.0 nM). Both compounds also had antioxidant properties. All compounds showed low in vitro metabolic stability, the only exception being compound 9b, which might be suitable for studies in vivo.
Collapse
|
6
|
Lanham KA, Nedden ML, Wise VE, Taylor MR. Genetically inducible and reversible zebrafish model of systemic inflammation. Biol Open 2022; 11:274172. [PMID: 35099005 PMCID: PMC8918989 DOI: 10.1242/bio.058559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022] Open
Abstract
The inflammatory response is a vital defense mechanism against trauma and pathogen induced damage, but equally important is its appropriate resolution. In some instances of severe trauma or sustained infection, inappropriate and persistent activation of the immune response can occur resulting in a dangerous systemic inflammatory response. Untreated, this systemic inflammatory response can lead to tissue damage, organ shutdown, and death. Replicating this condition in tractable model organisms can provide insight into the mechanisms involved in the induction, maintenance, and resolution of inflammation. To that end, we developed a non-invasive, inducible, and reversible model of systemic inflammation in zebrafish. Using the Gal4-EcR/UAS system activated by the ecdysone analog tebufenozide, we generated transgenic zebrafish that allow for chemically-induced, ubiquitous secretion of the mature form of zebrafish interleukin-1β (Il-1βmat) in both larval and adult developmental stages. To ensure a robust immune response, we attached a strong signal peptide from the Gaussia princeps luciferase enzyme to promote active secretion of the cytokine. We observe a dose-dependent inflammatory response involving neutrophil expansion accompanied by tissue damage and reduced survival. Washout of tebufenozide permits inflammation resolution. We also establish the utility of this model for the identification of small molecule anti-inflammatory compounds by treatment with the immunosuppressant rapamycin. Taken together, these features make this model a valuable new tool that can aid in identifying potential new therapies while broadening our understanding of systemic inflammation, its impact on the immune system and its resolution.
Collapse
Affiliation(s)
- Kevin A Lanham
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Megan L Nedden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Virginia E Wise
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Michael R Taylor
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
7
|
Bhagavatheeswaran S, Ramachandran V, Shanmugam S, Balakrishnan A. Isopimpinellin extends antiangiogenic effect through overexpression of miR-15b-5p and downregulating angiogenic stimulators. Mol Biol Rep 2021; 49:279-291. [PMID: 34709570 DOI: 10.1007/s11033-021-06870-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Angiogenesis is the formation of new blood vessels from an existing vasculature through a series of processes such as activation, proliferation, and directed migration of endothelial cells. Angiogenesis is instrumental in the metastatic spread of tumors. Isopimpinellin, a furanocoumarin group of phytochemicals, is an anticarcinogenic agent. However, no studies have proven its antiangiogenic effects. The current study thus aimed to screen the antiangiogenic effect of isopimpinellin. METHODS AND RESULTS Human Umblical Vein Endothelial Cell (HUVEC) as an in vitro model and zebrafish embryos as an in vivo model was used in this study. The experimental results showed that isopimpinellin effectively inhibited HUVEC proliferation, invasion, migration, and tube formation, which are the key steps in angiogenesis by markedly suppressing the expression of pro-angiogenic genes VEGF, AKT, and HIF-1α. In addition, isopimpinellin exerts its anti-angiogenic effect through the regulation of miR-15b-5p and miR-542-3p. Furthermore, in zebrafish embryos, isopimpinellin inhibited the development of intersegmental vessels (ISVs) through the significant downregulation of all pro-angiogenic genes vegf, vegfr2, survivin, angpt-1, angpt-2, and tie-2. CONCLUSION Collectively, these experimental findings offer novel insights into the antiangiogenic nature of isopimpinellin and open new avenues for therapeutic approaches.
Collapse
Affiliation(s)
| | - Vinu Ramachandran
- Department of Genetics, Dr. ALM PG IBMS, University of Madras, Chennai, Tamilnadu, 600113, India
| | - Sambantham Shanmugam
- Department of Pharmacology and Neuro Science, Texas Tech University Health Sciences, Lubbock, TX, 79430, USA
| | - Anandan Balakrishnan
- Department of Genetics, Dr. ALM PG IBMS, University of Madras, Chennai, Tamilnadu, 600113, India.
| |
Collapse
|
8
|
Papies J, Emanuel J, Heinemann N, Kulić Ž, Schroeder S, Tenner B, Lehner MD, Seifert G, Müller MA. Antiviral and Immunomodulatory Effects of Pelargonium sidoides DC. Root Extract EPs® 7630 in SARS-CoV-2-Infected Human Lung Cells. Front Pharmacol 2021; 12:757666. [PMID: 34759825 PMCID: PMC8573200 DOI: 10.3389/fphar.2021.757666] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Treatment options for COVID-19 are currently limited. Drugs reducing both viral loads and SARS-CoV-2-induced inflammatory responses would be ideal candidates for COVID-19 therapeutics. Previous in vitro and clinical studies suggest that the proprietary Pelargonium sidoides DC. root extract EPs 7630 has antiviral and immunomodulatory properties, limiting symptom severity and disease duration of infections with several upper respiratory viruses. Here we assessed if EPs 7630 affects SARS-CoV-2 propagation and the innate immune response in the human lung cell line Calu-3. In direct comparison to other highly pathogenic CoV (SARS-CoV, MERS-CoV), SARS-CoV-2 growth was most efficiently inhibited at a non-toxic concentration with an IC50 of 1.61 μg/ml. Particularly, the cellular entry step of SARS-CoV-2 was significantly reduced by EPs 7630 pretreatment (10-100 μg/ml) as shown by spike protein-carrying pseudovirus particles and infectious SARS-CoV-2. Using sequential ultrafiltration, EPs 7630 was separated into fractions containing either prodelphinidins of different oligomerization degrees or small molecule constituents like benzopyranones and purine derivatives. Prodelphinidins with a low oligomerization degree and small molecule constituents were most efficient in inhibiting SARS-CoV-2 entry already at 10 μg/ml and had comparable effects on immune gene regulation as EPs 7630. Downregulation of multiple pro-inflammatory genes (CCL5, IL6, IL1B) was accompanied by upregulation of anti-inflammatory TNFAIP3 at 48 h post-infection. At high concentrations (100 μg/ml) moderately oligomerized prodelphinidins reduced SARS-CoV-2 propagation most efficiently and exhibited pronounced immune gene modulation. Assessment of cytokine secretion in EPs 7630-treated and SARS-CoV-2-coinfected Calu-3 cells showed that pro-inflammatory cytokines IL-1β and IL-6 were elevated whereas multiple other COVID-19-associated cytokines (IL-8, IL-13, TNF-α), chemokines (CXCL9, CXCL10), and growth factors (PDGF, VEGF-A, CD40L) were significantly reduced by EPs 7630. SARS-CoV-2 entry inhibition and the differential immunomodulatory functions of EPs 7630 against SARS-CoV-2 encourage further in vivo studies.
Collapse
Affiliation(s)
- Jan Papies
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Jackson Emanuel
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Nicolas Heinemann
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Žarko Kulić
- Preclinical R & D, Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Simon Schroeder
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Beate Tenner
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin D. Lehner
- Preclinical R & D, Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Georg Seifert
- Department of Paediatric Oncology/Haematology, Otto-Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Paediatrics, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Marcel A. Müller
- Institute of Virology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow, Russia
| |
Collapse
|
9
|
Isles HM, Loynes CA, Alasmari S, Kon FC, Henry KM, Kadochnikova A, Hales J, Muir CF, Keightley MC, Kadirkamanathan V, Hamilton N, Lieschke GJ, Renshaw SA, Elks PM. Pioneer neutrophils release chromatin within in vivo swarms. eLife 2021; 10:68755. [PMID: 34292151 PMCID: PMC8298094 DOI: 10.7554/elife.68755] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022] Open
Abstract
Neutrophils are rapidly recruited to inflammatory sites where their coordinated migration forms clusters, a process termed neutrophil swarming. The factors that modulate early stages of neutrophil swarming are not fully understood, requiring the development of new in vivo models. Using transgenic zebrafish larvae to study endogenous neutrophil migration in a tissue damage model, we demonstrate that neutrophil swarming is a conserved process in zebrafish immunity, sharing essential features with mammalian systems. We show that neutrophil swarms initially develop around an individual pioneer neutrophil. We observed the violent release of extracellular cytoplasmic and nuclear fragments by the pioneer and early swarming neutrophils. By combining in vitro and in vivo approaches to study essential components of neutrophil extracellular traps (NETs), we provide in-depth characterisation and high-resolution imaging of the composition and morphology of these release events. Using a photoconversion approach to track neutrophils within developing swarms, we identify that the fate of swarm-initiating pioneer neutrophils involves extracellular chromatin release and that the key NET components gasdermin, neutrophil elastase, and myeloperoxidase are required for the swarming process. Together our findings demonstrate that release of cellular components by pioneer neutrophils is an initial step in neutrophil swarming at sites of tissue injury.
Collapse
Affiliation(s)
- Hannah M Isles
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Catherine A Loynes
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Sultan Alasmari
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Fu Chuen Kon
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Katherine M Henry
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Anastasia Kadochnikova
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Jack Hales
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Clare F Muir
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | | | - Visakan Kadirkamanathan
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Noémie Hamilton
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Stephen A Renshaw
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Philip M Elks
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| |
Collapse
|
10
|
Dong R, Tian Q, Shi Y, Chen S, Zhang Y, Deng Z, Wang X, Yao Q, Han L. An Integrated Strategy for Rapid Discovery and Identification of Quality Markers in Gardenia Fructus Using an Omics Discrimination-Grey Correlation-Biological Verification Method. Front Pharmacol 2021; 12:705498. [PMID: 34248647 PMCID: PMC8264552 DOI: 10.3389/fphar.2021.705498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 01/10/2023] Open
Abstract
Background: Gardenia Fructus (GF), a traditional Chinese medicine of Gardenia Ellis in Rubiaceae family, has the potential to clear heat and purge fire and has been widely used to treat multiple infection-related diseases. However, the quality markers (Q-Markers) of GF have not been revealed comprehensively. Methods: In this experiment, the transgenic zebrafish lines, Tg (l-fabp:EGFP) and Tg (lyz:EGFP), were used to evaluate two main kinds of traditional efficacies of GF, hepatoprotective and anti-inflammatory effects. All the GF samples from different production areas were tested their anti-liver injury and anti-inflammantory activities. High-performance liquid chromatography-quadrupole time-of-flight mass spectrometry method (HPLC-Q-TOF/MS) was employed for herbal metabonomic analysis of GF samples. Gray correlation analysis (GCA) was utilized to screen out the components closely associated with the activities. Finally, the zebrafish model was applied to verify the bioactivity of the crucial components to determine the Q-Markers of GF. Results: The zebrafish models were established by inducing with hydrogen peroxide or copper sulfate and applied to quickly evaluate the hepatoprotective effect and inflammation of GF samples. 27 potentially active components for liver protection and 21 potentially active components with anti-inflammatory properties were identified by herbal metabolomic analysis based on HPLC-Q-TOF/MS. The GCA result showed that five of the 27 components were highly correlated with liver protection, 15 of the 21 components were highly correlated with anti-inflammatory activity. Among them, geniposide and crocin-1 were confirmed their bioactivities on zebrafish experiment to be responsible for the protective effects of GF against liver injury, and genipin-1-β-D-gentiobioside, quinic acid, gardenoside, d-glucuronic acid, l-malic acid, mannitol, rutin, and chlorogenic acid were confirmed to be responsible for the anti-inflammatory effects. Finally, according to the screening principles of Q-Markers, genipin-1-β-D-gentiobioside, geniposide, and gardenoside were preliminarily identified to be the Q-Markers of GF. Conclusion: This study established an effective research strategy of “Omics Discrimination-Grey Correlation-Biological Verification,” which enabled the rapid identification of key pharmacological components of GF. These markers have provided a scientific basis for constructing a modern quality evaluation system for GF.
Collapse
Affiliation(s)
- Rong Dong
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qingping Tian
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, China
| | - Yongping Shi
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China.,School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, China.,Taiyuan Maternity and Child Health Care Hospital, Taiyuan, China
| | - Shanjun Chen
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yougang Zhang
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China.,School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, China
| | - Zhipeng Deng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qingqiang Yao
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liwen Han
- School of Pharmacy and Pharmaceutical Science, Shandong Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
11
|
Xie Y, Meijer AH, Schaaf MJM. Modeling Inflammation in Zebrafish for the Development of Anti-inflammatory Drugs. Front Cell Dev Biol 2021; 8:620984. [PMID: 33520995 PMCID: PMC7843790 DOI: 10.3389/fcell.2020.620984] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the inflammatory response in humans can lead to various inflammatory diseases, like asthma and rheumatoid arthritis. The innate branch of the immune system, including macrophage and neutrophil functions, plays a critical role in all inflammatory diseases. This part of the immune system is well-conserved between humans and the zebrafish, which has emerged as a powerful animal model for inflammation, because it offers the possibility to image and study inflammatory responses in vivo at the early life stages. This review focuses on different inflammation models established in zebrafish, and how they are being used for the development of novel anti-inflammatory drugs. The most commonly used model is the tail fin amputation model, in which part of the tail fin of a zebrafish larva is clipped. This model has been used to study fundamental aspects of the inflammatory response, like the role of specific signaling pathways, the migration of leukocytes, and the interaction between different immune cells, and has also been used to screen libraries of natural compounds, approved drugs, and well-characterized pathway inhibitors. In other models the inflammation is induced by chemical treatment, such as lipopolysaccharide (LPS), leukotriene B4 (LTB4), and copper, and some chemical-induced models, such as treatment with trinitrobenzene sulfonic acid (TNBS), specifically model inflammation in the gastro-intestinal tract. Two mutant zebrafish lines, carrying a mutation in the hepatocyte growth factor activator inhibitor 1a gene (hai1a) and the cdp-diacylglycerolinositol 3-phosphatidyltransferase (cdipt) gene, show an inflammatory phenotype, and they provide interesting model systems for studying inflammation. These zebrafish inflammation models are often used to study the anti-inflammatory effects of glucocorticoids, to increase our understanding of the mechanism of action of this class of drugs and to develop novel glucocorticoid drugs. In this review, an overview is provided of the available inflammation models in zebrafish, and how they are used to unravel molecular mechanisms underlying the inflammatory response and to screen for novel anti-inflammatory drugs.
Collapse
|
12
|
Foulkes MJ, Tolliday FH, Henry KM, Renshaw SA, Jones S. Evaluation of the anti-inflammatory effects of synthesised tanshinone I and isotanshinone I analogues in zebrafish. PLoS One 2020; 15:e0240231. [PMID: 33022012 PMCID: PMC7537861 DOI: 10.1371/journal.pone.0240231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/22/2020] [Indexed: 01/13/2023] Open
Abstract
During inflammation, dysregulated neutrophil behaviour can play a major role in a range of chronic inflammatory diseases, for many of which current treatments are generally ineffective. Recently, specific naturally occurring tanshinones have shown promising anti-inflammatory effects by targeting neutrophils in vivo, yet such tanshinones, and moreover, their isomeric isotanshinone counterparts, are still a largely underexplored class of compounds, both in terms of synthesis and biological effects. To explore the anti-inflammatory effects of isotanshinones, and the tanshinones more generally, a series of substituted tanshinone and isotanshinone analogues was synthesised, alongside other structurally similar molecules. Evaluation of these using a transgenic zebrafish model of neutrophilic inflammation revealed differential anti-inflammatory profiles in vivo, with a number of compounds exhibiting promising effects. Several compounds reduce initial neutrophil recruitment and/or promote resolution of neutrophilic inflammation, of which two also result in increased apoptosis of human neutrophils. In particular, the methoxy-substituted tanshinone 39 specifically accelerates resolution of inflammation without affecting the recruitment of neutrophils to inflammatory sites, making this a particularly attractive candidate for potential pro-resolution therapeutics, as well as a possible lead for future development of functionalised tanshinones as molecular tools and/or chemical probes. The structurally related β-lapachones promote neutrophil recruitment but do not affect resolution. We also observed notable differences in toxicity profiles between compound classes. Overall, we provide new insights into the in vivo anti-inflammatory activities of several novel tanshinones, isotanshinones, and structurally related compounds.
Collapse
Affiliation(s)
- Matthew J. Foulkes
- Department of Chemistry, The University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Faith H. Tolliday
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Katherine M. Henry
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- The Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Simon Jones
- Department of Chemistry, The University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Zanandrea R, Bonan CD, Campos MM. Zebrafish as a model for inflammation and drug discovery. Drug Discov Today 2020; 25:2201-2211. [PMID: 33035664 DOI: 10.1016/j.drudis.2020.09.036] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/17/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022]
Abstract
Zebrafish is a small teleost (bony) fish used in many areas of pharmacology and toxicology. This animal model has advantages for the discovery of anti-inflammatory drugs, such as the potential for real-time assessment of cell migration mechanisms. Additionally, zebrafish display a repertoire of inflammatory cells, mediators, and receptors that are similar to those in mammals, including humans. Inflammatory disease modeling in either larvae or adult zebrafish represents a promising tool for the screening of new anti-inflammatory compounds, contributing to our understanding of the mechanisms involved in chronic inflammatory conditions. In this review, we provide an overview of the characterization of inflammatory responses in zebrafish, emphasizing its relevance for drug discovery in this research area.
Collapse
Affiliation(s)
- Rodrigo Zanandrea
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil
| | - Carla D Bonan
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Porto Alegre, RS, Brazil
| | - Maria M Campos
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Centro de Pesquisa em Toxicologia e Farmacologia, Porto Alegre, RS, Brazil.
| |
Collapse
|
14
|
Isoniazid promotes the anti-inflammatory response in zebrafish associated with regulation of the PPARγ/NF-κB/AP-1 pathway. Chem Biol Interact 2020; 316:108928. [DOI: 10.1016/j.cbi.2019.108928] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 12/15/2019] [Indexed: 12/26/2022]
|
15
|
Vaz RL, Sousa S, Chapela D, van der Linde HC, Willemsen R, Correia AD, Outeiro TF, Afonso ND. Identification of antiparkinsonian drugs in the 6-hydroxydopamine zebrafish model. Pharmacol Biochem Behav 2019; 189:172828. [PMID: 31785245 DOI: 10.1016/j.pbb.2019.172828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/09/2023]
Abstract
Parkinson's disease (PD) is known as a movement disorder due to characteristic motor features. Existing therapies for PD are only symptomatic, and their efficacy decreases as disease progresses. Zebrafish, a vertebrate in which parkinsonism has been modelled, offers unique features for the identification of molecules with antiparkinsonian properties. Here, we developed a screening assay for the selection of neuroactive agents with antiparkinsonian potential. First, we performed a pharmacological validation of the phenotypes exhibited by the 6-hydroxydopamine zebrafish model, by testing the effects of known antiparkinsonian agents. These drugs were also tested for disease-modifying properties by whole mount immunohistochemistry to TH+ neurons and confocal microscopy in the dopaminergic diencephalic cluster of zebrafish. Next, we optimized a phenotypic screening using the 6-hydroxydopamine zebrafish model and tested 1600 FDA-approved bioactive drugs. We found that 6-hydroxydopamine-lesioned zebrafish larvae exhibit bradykinetic and dyskinetic-like behaviours that are rescued by the administration of levodopa, rasagiline, isradipine or amantadine. The rescue of dopaminergic cell loss by isradipine was also verified, through the observation of a higher number of TH+ neurons in 6-OHDA-lesioned zebrafish larvae treated with this compound as compared to untreated lesioned larvae. The phenotypic screening enabled us to identify several compounds previously positioned for PD, as well as, new molecules with potential antiparkinsonian properties. Among these, we selected stavudine, tapentadol and nabumetone as the most promising candidates. Our results demonstrate the functional similarities of the motor impairments exhibited by 6-hydroxydopamine-lesioned zebrafish with mammalian models of PD and with PD patients, and highlights novel molecules with antiparkinsonian potential.
Collapse
Affiliation(s)
- Rita L Vaz
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Sousa
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.
| | - Diana Chapela
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | - Rob Willemsen
- Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Ana D Correia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; CEDOC, Chronic Diseases Research Centre, NOVA Medical School |Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal.; Max Planck Institute for Experimental Medicine, Göttingen, Germany; Institute of Neuroscience, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Nuno D Afonso
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
16
|
Isles HM, Herman KD, Robertson AL, Loynes CA, Prince LR, Elks PM, Renshaw SA. The CXCL12/CXCR4 Signaling Axis Retains Neutrophils at Inflammatory Sites in Zebrafish. Front Immunol 2019; 10:1784. [PMID: 31417560 PMCID: PMC6684839 DOI: 10.3389/fimmu.2019.01784] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/15/2019] [Indexed: 12/23/2022] Open
Abstract
The inappropriate retention of neutrophils at inflammatory sites is a major driver of the excessive tissue damage characteristic of respiratory inflammatory diseases including COPD, ARDS, and cystic fibrosis. The molecular programmes which orchestrate neutrophil recruitment to inflammatory sites through chemotactic guidance have been well-studied. However, how neutrophil sensitivity to these cues is modulated during inflammation resolution is not understood. The identification of neutrophil reverse migration as a mechanism of inflammation resolution and the ability to modulate this therapeutically has identified a new target to treat inflammatory disease. Here we investigate the role of the CXCL12/CXCR4 signaling axis in modulating neutrophil retention at inflammatory sites. We used an in vivo tissue injury model to study neutrophilic inflammation using transgenic zebrafish larvae. Expression of cxcl12a and cxcr4b during the tissue damage response was assessed using in situ hybridization and analysis of RNA sequencing data. CRISPR/Cas9 was used to knockdown cxcl12a and cxcr4b in zebrafish larvae. The CXCR4 antagonist AMD3100 was used to block the Cxcl12/Cxcr4 signaling axis pharmacologically. We identified that cxcr4b and cxcl12a are expressed at the wound site in zebrafish larvae during the inflammatory response. Following tail-fin transection, removal of neutrophils from inflammatory sites is significantly increased in cxcr4b and cxcl12a CRISPR knockdown larvae. Pharmacological inhibition of the Cxcl12/Cxcr4 signaling axis accelerated resolution of the neutrophil component of inflammation, an effect caused by an increase in neutrophil reverse migration. The findings of this study suggest that CXCR4/CXCL12 signaling may play an important role in neutrophil retention at inflammatory sites, identifying a potential new target for the therapeutic removal of neutrophils from the lung in chronic inflammatory disease.
Collapse
Affiliation(s)
- Hannah M. Isles
- Renshaw Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Elks Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Kimberly D. Herman
- Renshaw Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Prince Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Anne L. Robertson
- Renshaw Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Zon Laboratory, Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Catherine A. Loynes
- Renshaw Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Lynne R. Prince
- Prince Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Philip M. Elks
- Elks Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Renshaw Laboratory, Department of Infection, Immunity, and Cardiovascular Disease, The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
17
|
Xie Y, Tolmeijer S, Oskam JM, Tonkens T, Meijer AH, Schaaf MJM. Glucocorticoids inhibit macrophage differentiation towards a pro-inflammatory phenotype upon wounding without affecting their migration. Dis Model Mech 2019; 12:dmm.037887. [PMID: 31072958 PMCID: PMC6550045 DOI: 10.1242/dmm.037887] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/24/2019] [Indexed: 12/18/2022] Open
Abstract
Glucocorticoid drugs are widely used to treat immune-related diseases, but their use is limited by side effects and by resistance, which especially occurs in macrophage-dominated diseases. In order to improve glucocorticoid therapies, more research is required into the mechanisms of glucocorticoid action. In the present study, we have used a zebrafish model for inflammation to study glucocorticoid effects on the innate immune response. In zebrafish larvae, the migration of neutrophils towards a site of injury is inhibited upon glucocorticoid treatment, whereas migration of macrophages is glucocorticoid resistant. We show that wounding-induced increases in the expression of genes that encode neutrophil-specific chemoattractants (Il8 and Cxcl18b) are attenuated by the synthetic glucocorticoid beclomethasone, but that beclomethasone does not attenuate the induction of the genes encoding Ccl2 and Cxcl11aa, which are required for macrophage recruitment. RNA sequencing on FACS-sorted macrophages shows that the vast majority of the wounding-induced transcriptional changes in these cells are inhibited by beclomethasone, whereas only a small subset is glucocorticoid-insensitive. As a result, beclomethasone decreases the number of macrophages that differentiate towards a pro-inflammatory (M1) phenotype, which we demonstrated using a tnfa:eGFP-F reporter line and analysis of macrophage morphology. We conclude that differentiation and migration of macrophages are regulated independently, and that glucocorticoids leave the chemotactic migration of macrophages unaffected, but exert their anti-inflammatory effect on these cells by inhibiting their differentiation to an M1 phenotype. The resistance of macrophage-dominated diseases to glucocorticoid therapy can therefore not be attributed to an intrinsic insensitivity of macrophages to glucocorticoids. Summary: In a zebrafish model for inflammation, glucocorticoids do not affect the migration of macrophages, but inhibit their differentiation towards an M1 phenotype, by strongly attenuating transcriptional responses in these cells.
Collapse
Affiliation(s)
- Yufei Xie
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| | - Sofie Tolmeijer
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| | - Jelle M Oskam
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| | - Tijs Tonkens
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| | - Annemarie H Meijer
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| | - Marcel J M Schaaf
- Animal Science and Health Cluster, Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands
| |
Collapse
|
18
|
Vaz RL, Outeiro TF, Ferreira JJ. Zebrafish as an Animal Model for Drug Discovery in Parkinson's Disease and Other Movement Disorders: A Systematic Review. Front Neurol 2018; 9:347. [PMID: 29910763 PMCID: PMC5992294 DOI: 10.3389/fneur.2018.00347] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Movement disorders can be primarily divided into hypokinetic and hyperkinetic. Most of the hypokinetic syndromes are associated with the neurodegenerative disorder Parkinson’s disease (PD). By contrast, hyperkinetic syndromes encompass a broader array of diseases, including dystonia, essential tremor, or Huntington’s disease. The discovery of effective therapies for these disorders has been challenging and has also involved the development and characterization of accurate animal models for the screening of new drugs. Zebrafish constitutes an alternative vertebrate model for the study of movement disorders. The neuronal circuitries involved in movement in zebrafish are well characterized, and most of the associated molecular mechanisms are highly conserved. Particularly, zebrafish models of PD have contributed to a better understanding of the role of several genes implicated in the disease. Furthermore, zebrafish is a vertebrate model particularly suited for large-scale drug screenings. The relatively small size of zebrafish, optical transparency, and lifecycle, are key characteristics that facilitate the study of multiple compounds at the same time. Several transgenic, knockdown, and mutant zebrafish lines have been generated and characterized. Therefore, it is central to critically analyze these zebrafish lines and understand their suitability as models of movement disorders. Here, we revise the pathogenic mechanisms, phenotypes, and responsiveness to pharmacotherapies of zebrafish lines of the most common movement disorders. A systematic review of the literature was conducted by including all studies reporting the characterization of zebrafish models of the movement disorders selected from five bibliographic databases. A total of 63 studies were analyzed, and the most relevant data within the scope of this review were gathered. The majority (62%) of the studies were focused in the characterization of zebrafish models of PD. Overall, the zebrafish models included display conserved biochemical and neurobehavioral features of the phenomenology in humans. Nevertheless, in light of what is known for all animal models available, the use of zebrafish as a model for drug discovery requires further optimization. Future technological developments alongside with a deeper understanding of the molecular bases of these disorders should enable the development of novel zebrafish lines that can prove useful for drug discovery for movement disorders.
Collapse
Affiliation(s)
- Rita L Vaz
- TechnoPhage, SA, Lisboa, Portugal.,Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany.,Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,CEDOC, Chronic Diseases Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,The Medical School, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joaquim J Ferreira
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.,Laboratory of Clinical Pharmacology and Therapeutics, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,CNS-Campus Neurológico Sénior, Torres Vedras, Portugal
| |
Collapse
|
19
|
Johnston HJ, Verdon R, Gillies S, Brown DM, Fernandes TF, Henry TB, Rossi AG, Tran L, Tucker C, Tyler CR, Stone V. Adoption of in vitro systems and zebrafish embryos as alternative models for reducing rodent use in assessments of immunological and oxidative stress responses to nanomaterials. Crit Rev Toxicol 2017; 48:252-271. [PMID: 29239234 DOI: 10.1080/10408444.2017.1404965] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Assessing the safety of engineered nanomaterials (NMs) is paramount to the responsible and sustainable development of nanotechnology, which provides huge societal benefits. Currently, there is no evidence that engineered NMs cause detrimental health effects in humans. However, investigation of NM toxicity using in vivo, in vitro, in chemico, and in silico models has demonstrated that some NMs stimulate oxidative stress and inflammation, which may lead to adverse health effects. Accordingly, investigation of these responses currently dominates NM safety assessments. There is a need to reduce reliance on rodent testing in nanotoxicology for ethical, financial and legislative reasons, and due to evidence that rodent models do not always predict the human response. We advocate that in vitro models and zebrafish embryos should have greater prominence in screening for NM safety, to better align nanotoxicology with the 3Rs principles. Zebrafish are accepted for use by regulatory agencies in chemical safety assessments (e.g. developmental biology) and there is growing acceptance of their use in biomedical research, providing strong foundations for their use in nanotoxicology. We suggest that investigation of the response of phagocytic cells (e.g. neutrophils, macrophages) in vitro should also form a key part of NM safety assessments, due to their prominent role in the first line of defense. The development of a tiered testing strategy for NM hazard assessment that promotes the more widespread adoption of non-rodent, alternative models and focuses on investigation of inflammation and oxidative stress could make nanotoxicology testing more ethical, relevant, and cost and time efficient.
Collapse
Affiliation(s)
| | - Rachel Verdon
- a Nano Safety Research Group , Heriot-Watt University , Edinburgh , UK
| | - Suzanne Gillies
- a Nano Safety Research Group , Heriot-Watt University , Edinburgh , UK
| | - David M Brown
- a Nano Safety Research Group , Heriot-Watt University , Edinburgh , UK
| | | | - Theodore B Henry
- a Nano Safety Research Group , Heriot-Watt University , Edinburgh , UK
| | - Adriano G Rossi
- b Medical Research Council (MRC) Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh , Edinburgh , UK
| | - Lang Tran
- c Institute of Occupational Medicine , Edinburgh , UK
| | - Carl Tucker
- b Medical Research Council (MRC) Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh , Edinburgh , UK
| | - Charles R Tyler
- d Department of Biosciences , College of Life and Environmental Sciences, University of Exeter , Exeter , UK
| | - Vicki Stone
- a Nano Safety Research Group , Heriot-Watt University , Edinburgh , UK
| |
Collapse
|
20
|
Foulkes MJ, Henry KM, Rougeot J, Hooper-Greenhill E, Loynes CA, Jeffrey P, Fleming A, Savage CO, Meijer AH, Jones S, Renshaw SA. Expression and regulation of drug transporters in vertebrate neutrophils. Sci Rep 2017; 7:4967. [PMID: 28694436 PMCID: PMC5504015 DOI: 10.1038/s41598-017-04785-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/19/2017] [Indexed: 12/16/2022] Open
Abstract
There remains a need to identify novel pro-resolution drugs for treatment of inflammatory disease. To date, there are no neutrophil-specific anti-inflammatory treatments in clinical use, perhaps due to our lack of understanding of how drugs access this complex cell type. Here we present the first comprehensive description and expression of both major classes of drug transporters, SLC and ABC, in resting human blood neutrophils. Moreover, we have studied the expression of these carriers in the tractable model system, the zebrafish (Danio rerio), additionally examining the evolutionary relationship between drug transporters in zebrafish and humans. We anticipate that this will be a valuable resource to the field of inflammation biology and will be an important asset in future anti-inflammatory drug design.
Collapse
Affiliation(s)
- Matthew J Foulkes
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Chemistry, The University of Sheffield, Sheffield, S3 7HF, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Katherine M Henry
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Julien Rougeot
- Institute of Biology, University of Leiden, 2333 CC, Leiden, The Netherlands
| | - Edward Hooper-Greenhill
- Immuno-Inflammation Therapy Area Unit, GlaxoSmithKline Research and Development Ltd., Stevenage, SG1 2NY, UK
| | - Catherine A Loynes
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Phil Jeffrey
- Rare Diseases Research Unit, Pfizer Ltd., Cambridge, CB21 6GP, UK
| | - Angeleen Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, CB2 2XY, UK
| | - Caroline O Savage
- Immuno-Inflammation Therapy Area Unit, GlaxoSmithKline Research and Development Ltd., Stevenage, SG1 2NY, UK
| | - Annemarie H Meijer
- Institute of Biology, University of Leiden, 2333 CC, Leiden, The Netherlands
| | - Simon Jones
- Department of Chemistry, The University of Sheffield, Sheffield, S3 7HF, UK
| | - Stephen A Renshaw
- The Bateson Centre, The University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, S10 2RX, UK.
| |
Collapse
|
21
|
Oehlers SH, Flores MV, Hall CJ, Wang L, Ko DC, Crosier KE, Crosier PS. A whole animal chemical screen approach to identify modifiers of intestinal neutrophilic inflammation. FEBS J 2017; 284:402-413. [PMID: 27885812 PMCID: PMC5296212 DOI: 10.1111/febs.13976] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/25/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022]
Abstract
By performing two high-content small molecule screens on dextran sodium sulfate- and trinitrobenzene sulfonic acid-induced zebrafish enterocolitis models of inflammatory bowel disease, we have identified novel anti-inflammatory drugs from the John Hopkins Clinical Compound Library that suppress neutrophilic inflammation. Live imaging of neutrophil distribution was used to assess the level of acute inflammation and concurrently screen for off-target drug effects. Supporting the validity of our screening strategy, most of the anti-inflammatory drug hits were known antibiotics or anti-inflammatory agents. Novel hits included cholecystokinin (CCK) and dopamine receptor agonists. Using a pharmacological approach, we show that while CCK and dopamine receptor agonists alleviate enterocolitis-associated inflammation, receptor antagonists exacerbate inflammation in zebrafish. This work highlights the utility of small molecule screening in zebrafish enterocolitis models as a tool to identify novel bioactive molecules capable of modulating acute inflammation.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Anti-Inflammatory Agents/pharmacology
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/pathology
- Crohn Disease/chemically induced
- Crohn Disease/drug therapy
- Crohn Disease/immunology
- Crohn Disease/pathology
- Dextran Sulfate
- Disease Models, Animal
- Dopamine Agonists/pharmacology
- Dysbiosis/chemically induced
- Dysbiosis/drug therapy
- Dysbiosis/immunology
- Dysbiosis/pathology
- Embryo, Nonmammalian
- Gene Expression
- High-Throughput Screening Assays
- Humans
- Immunologic Factors/pharmacology
- Intestines/drug effects
- Intestines/immunology
- Intestines/pathology
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/pathology
- Receptors, Cholecystokinin/agonists
- Receptors, Cholecystokinin/genetics
- Receptors, Cholecystokinin/immunology
- Receptors, Dopamine/genetics
- Receptors, Dopamine/immunology
- Small Molecule Libraries/pharmacology
- Trinitrobenzenesulfonic Acid
- Zebrafish
Collapse
Affiliation(s)
- Stefan H Oehlers
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019 Auckland Mail Centre, Auckland 1142, New Zealand
- Tuberculosis Research Program, Centenary Institute, Camperdown, NSW 2050, Australia
- Sydney Medical School, The University of Sydney, Newtown, Australia
| | - Maria Vega Flores
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019 Auckland Mail Centre, Auckland 1142, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019 Auckland Mail Centre, Auckland 1142, New Zealand
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University
- Department of Medicine, School of Medicine, Duke University
| | - Kathryn E Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019 Auckland Mail Centre, Auckland 1142, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Private Bag 92019 Auckland Mail Centre, Auckland 1142, New Zealand
| |
Collapse
|