1
|
Xiong Y, Shen T, Lou P, Yang J, Kastelic JP, Liu J, Xu C, Han B, Gao J. Colostrum-derived extracellular vesicles: potential multifunctional nanomedicine for alleviating mastitis. J Nanobiotechnology 2024; 22:627. [PMID: 39407245 PMCID: PMC11481564 DOI: 10.1186/s12951-024-02926-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Bovine mastitis is an infectious disease that causes substantial economic losses to the dairy industry worldwide. Current antibiotic therapy faces issues of antibiotic misuse and antimicrobial resistance, which has aroused concerns for both veterinary and human medicine. Thus, this study explored the potential of Colo EVs (bovine colostrum-derived extracellular vesicles) to address mastitis. Using LPS-induced murine mammary epithelial cells (HC11), mouse monocyte macrophages (RAW 264.7), and a murine mastitis model with BALB/C mice, we evaluated the safety and efficacy of Colo EVs, in vivo and in vitro. Colo EVs had favorable biosafety profiles, promoting cell proliferation and migration without inducing pathological changes after injection into murine mammary glands. In LPS-induced murine mastitis, Colo EVs significantly reduced inflammation, improved inflammatory scores, and preserved tight junction proteins while protecting milk production. Additionally, in vitro experiments demonstrated that Colo EVs downregulated inflammatory cytokine expression, reduced inflammatory markers, and attenuated NF-κB pathway activation. In summary, we inferred that Colo EVs have promise as a therapeutic approach for mastitis treatment, owing to their anti-inflammatory properties, potentially mediated through the NF-κB signaling pathway modulation.
Collapse
Affiliation(s)
- Yindi Xiong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Taiyu Shen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Peng Lou
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, 610213, Chengdu, China
| | - Jingyue Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - John P Kastelic
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, 610213, Chengdu, China
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
2
|
Lin X, Zhao Z, Cai Y, He Y, Wang J, Liu N, Qin Y, Wu Y. MyD88 deficiency in mammary epithelial cells attenuates lipopolysaccharide (LPS)-induced mastitis in mice. Biochem Biophys Res Commun 2024; 739:150569. [PMID: 39186869 DOI: 10.1016/j.bbrc.2024.150569] [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: 05/10/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Lactation mastitis is a debilitating inflammatory mammary disease in postpartum animals. Myeloid differentiation primary response protein MyD88 is the key downstream adapter for innate pattern recognition receptor toll-like receptor 4 (TLR4), which plays an important role in inflammation. However, the specific role of MyD88 in mammary epithelial cells in the progression of mastitis has not been investigated. In this study, lipopolysaccharide (LPS)-induced mouse mastitis model was used and cytokines such as Tnf-α, Il-1β, Il-6, Cxcl1, Cxcl2 and Ccl2 were significantly increased in inflammatory mammary gland as shown by real time-qPCR. However, the mice with MyD88-deficienet in mammary epithelial cells (cKO) showed a reduction in the expression of Tnf-α, Il-1β, Il-6, Cxcl1 and Cxcl2 in mammary gland compared with control mice, when subjected to LPS induced mastitis. Immunohistochemical staining of cleaved caspase-3 showed that the cell apoptosis induced by inflammation were decreased in MyD88 cKO mice. Furthermore, there were significantly fewer infiltrating inflammatory cells in alveolar lumen of MyD88 cKO mice, including Ly6G-positive neutrophils and F4/80-positive macrophages. RNA-seq in LPS treated mammary glands showed that MyD88 cKO mice had significantly downregulated inflammation-related genes and upregulated genes related to anti-inflammation processes and lipid metabolism compared with control mice. Thus, these results demonstrate that MyD88 in mammary epithelial cells is essential for mastitis progression. And this study not only has important implications for understanding the innate immune response in mammary epithelial cells, but also potentially helps the development of new therapeutic drugs for treating mastitis.
Collapse
Affiliation(s)
- Xinyi Lin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhifeng Zhao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuqing Cai
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yifeilong He
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jing Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
3
|
Jermann PM, Wagner LA, Fritsche D, Gross JJ, Wellnitz O, Bruckmaier RM. Acute phase reaction to lipopolysaccharide-induced mastitis in early lactation dairy cows fed nitrogenic, glucogenic, or lipogenic diets. J Dairy Sci 2023; 106:9879-9891. [PMID: 37678770 DOI: 10.3168/jds.2023-23582] [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: 04/05/2023] [Accepted: 07/11/2023] [Indexed: 09/09/2023]
Abstract
The availability of certain macronutrients is likely to influence the capacity of the immune system. Therefore, we investigated the acute phase response to intramammary (i.mam.) lipopolysaccharide (LPS) in dairy cows fed a nitrogenic diet (n = 10) high in crude protein, a glucogenic diet (n = 11) high in carbohydrates and glucogenic precursors, or a lipogenic diet (n = 11) high in lipids. Thirty-two dairy cows were fed one of the dietary concentrates directly after calving until the end of trial at 27 ± 3 days in milk (mean ± standard deviation). In wk 3 of lactation, 20 µg of LPS was i.mam. injected in one quarter, and sterile NaCl (0.9%) in the contralateral quarter. Milk samples of the LPS-challenged and control quarter were taken hourly from before (0 h) until 9 h after LPS challenge and analyzed for milk amyloid A (MAA), haptoglobin (HP), and IL-8. In addition, blood samples were taken in the morning, and composite milk samples at morning and evening milkings, from 1 d before until 3 d after LPS challenge, and again on d 9, to determine serum amyloid A (SAA) and HP in blood, and MAA and HP in milk. The mRNA abundance of various immunological and metabolic factors in blood leukocytes was quantified by quantitative reverse-transcription PCR from samples taken at -18, -1, 6, 9, and 23 h relative to LPS application. The dietary concentrates did not affect any of the parameters in blood, milk, and leukocytes. The IL-8 was increased from 2 h, HP from 2 to 3 h, and MAA from 6 h relative to the LPS administration in the milk of the challenged quarter and remained elevated until 9 h. The MAA and HP were also increased at 9 h after LPS challenge in whole-udder composite milk, whereas HP and SAA in blood were increased only after 23 h. All 4 parameters were decreased again on d 9. Similar for all groups, the mRNA abundance of HP and the heat shock protein family A increased after the LPS challenge, whereas the mRNA expression of the tumor necrosis factor α and the leukocyte integrin β 2 subunit (CD18) were decreased at 6 h after LPS challenge. The glucose transporter (GLUT)1 mRNA abundance decreased after LPS, whereas that of the GLUT3 increased, and that of the GLUT4 was not detectable. The mRNA abundance of GAPDH was increased at 9 h after LPS and remained elevated. The acute phase protein response was detected earlier in milk compared with blood indicating mammary production. However, immunological responses to LPS were not affected by the availability of specific macronutrients provided by the different diets.
Collapse
Affiliation(s)
- P M Jermann
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - L A Wagner
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - D Fritsche
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - J J Gross
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - O Wellnitz
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - R M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|
4
|
Li M, Wang Z, Fu S, Sun N, Li W, Xu Y, Han X, Zhang J, Miao J. Taurine reduction of injury from neutrophil infiltration ameliorates Streptococcus uberis-induced mastitis. Int Immunopharmacol 2023; 124:111028. [PMID: 37857121 DOI: 10.1016/j.intimp.2023.111028] [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: 08/01/2023] [Revised: 09/14/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023]
Abstract
Mastitis is a common disease of dairy cows characterized by infiltration of leukocytes, especially neutrophils, resulting in increased permeability of the blood-milk barrier (BMB). Taurine, a functional nutrient, has been shown to have anti-inflammatory and antioxidant effects. Here, we investigated the regulatory effects and mechanisms of taurine on the complex immune network of the mammary gland in Streptococcus uberis (S. uberis) infection. We found that taurine had no direct effect on CXCL2-mediated neutrophil chemotaxis. However, it inhibited MAPK and NF-κB signalings by modulating the activity of TAK1 downstream of TLR2, thereby reducing CXCL2 expression in macrophages to reduce neutrophil recruitment in S. uberis infection. Further, the AMPK/Nrf2 signaling pathway was activated by taurine to help mitigate oxidative damage, apoptosis and disruption of tight junctions in mammary epithelial cells caused by hypochlorous acid, a strong oxidant produced by neutrophils, thus protecting the integrity of the mammary epithelial barrier. Taurine protects the BMB from damage caused by neutrophils via blocking the macrophage-CXCL2-neutrophil signaling axis and increasing the antioxidant capacity of mammary epithelial cells.
Collapse
Affiliation(s)
- Ming Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Zhenglei Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Naiyan Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Weizhen Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jinqiu Zhang
- National Research Center for Veterinary Vaccine Engineering and Technology of China, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Ji ZH, Gao F, Xie WY, Wu HY, Ren WZ, Yuan B. Mammary Epithelial Cell-Derived Exosomal miR-221-3p Regulates Macrophage Polarization by Targeting Igf2 bp2 during Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14742-14757. [PMID: 37757458 DOI: 10.1021/acs.jafc.3c03350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Mastitis affects the milk quality and yield and is the most expensive disease in dairy cows. Elucidation of the pathogenesis of mastitis is of great importance for disease control. As a medium of intercellular communication, exosomes play key roles in various inflammatory diseases by regulating macrophage polarization. However, the molecular factors in exosomes that mediate the intercellular communication between mammary epithelial cells and macrophages during mastitis remain to be further explored. In this study, we isolated and identified mammary epithelial cell-derived exosomes from a lipopolysaccharide (LPS)/lipoteichoic acid (LTA)-induced mastitis cell model, and we demonstrated that exosomes from LPS/LTA-stimulated mammary epithelial cells promote M1-type macrophage polarization in vivo and in vitro. Based on the results of high-throughput sequencing, we constructed a differential miRNA (microRNA) expression profile of exosomes and demonstrated that miR-221-3p was highly expressed. Furthermore, in vivo and in vitro experiments, combined with coculture experiments and fluorescence tracing, showed that high miR-221-3p expression promoted M1-type macrophage polarization, demonstrating the transcellular role of miR-221-3p. Mechanistically, dual luciferase reporter gene assays and rescue assays showed that miR-221-3p regulated macrophage polarization by targeting Igf2bp2. The results of this study will deepen our understanding of the pathogenesis of mastitis, and the molecular regulatory axis that was established in this study is expected to be a target for mastitis treatment.
Collapse
Affiliation(s)
- Zhong-Hao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| | - Wen-Yin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| | - Hong-Yu Wu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
- Jilin Academy of Agricultural Sciences, Jilin 132101, China
| | - Wen-Zhi Ren
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| |
Collapse
|
6
|
Hasankhani A, Bakherad M, Bahrami A, Shahrbabak HM, Pecho RDC, Shahrbabak MM. Integrated analysis of inflammatory mRNAs, miRNAs, and lncRNAs elucidates the molecular interactome behind bovine mastitis. Sci Rep 2023; 13:13826. [PMID: 37620551 PMCID: PMC10449796 DOI: 10.1038/s41598-023-41116-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023] Open
Abstract
Mastitis is known as intramammary inflammation, which has a multifactorial complex phenotype. However, the underlying molecular pathogenesis of mastitis remains poorly understood. In this study, we utilized a combination of RNA-seq and miRNA-seq techniques, along with computational systems biology approaches, to gain a deeper understanding of the molecular interactome involved in mastitis. We retrieved and processed one hundred transcriptomic libraries, consisting of 50 RNA-seq and 50 matched miRNA-seq data, obtained from milk-isolated monocytes of Holstein-Friesian cows, both infected with Streptococcus uberis and non-infected controls. Using the weighted gene co-expression network analysis (WGCNA) approach, we constructed co-expressed RNA-seq-based and miRNA-seq-based modules separately. Module-trait relationship analysis was then performed on the RNA-seq-based modules to identify highly-correlated modules associated with clinical traits of mastitis. Functional enrichment analysis was conducted to understand the functional behavior of these modules. Additionally, we assigned the RNA-seq-based modules to the miRNA-seq-based modules and constructed an integrated regulatory network based on the modules of interest. To enhance the reliability of our findings, we conducted further analyses, including hub RNA detection, protein-protein interaction (PPI) network construction, screening of hub-hub RNAs, and target prediction analysis on the detected modules. We identified a total of 17 RNA-seq-based modules and 3 miRNA-seq-based modules. Among the significant highly-correlated RNA-seq-based modules, six modules showed strong associations with clinical characteristics of mastitis. Functional enrichment analysis revealed that the turquoise module was directly related to inflammation persistence and mastitis development. Furthermore, module assignment analysis demonstrated that the blue miRNA-seq-based module post-transcriptionally regulates the turquoise RNA-seq-based module. We also identified a set of different RNAs, including hub-hub genes, hub-hub TFs (transcription factors), hub-hub lncRNAs (long non-coding RNAs), and hub miRNAs within the modules of interest, indicating their central role in the molecular interactome underlying the pathogenic mechanisms of S. uberis infection. This study provides a comprehensive insight into the molecular crosstalk between immunoregulatory mRNAs, miRNAs, and lncRNAs during S. uberis infection. These findings offer valuable directions for the development of molecular diagnosis and biological therapies for mastitis.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | - Maryam Bakherad
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | - Hossein Moradi Shahrbabak
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | | | - Mohammad Moradi Shahrbabak
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| |
Collapse
|
7
|
Cansever D, Petrova E, Krishnarajah S, Mussak C, Welsh CA, Mildenberger W, Mulder K, Kreiner V, Roussel E, Stifter SA, Andreadou M, Zwicky P, Jurado NP, Rehrauer H, Tan G, Liu Z, Blériot C, Ronchi F, Macpherson AJ, Ginhoux F, Natalucci G, Becher B, Greter M. Lactation-associated macrophages exist in murine mammary tissue and human milk. Nat Immunol 2023:10.1038/s41590-023-01530-0. [PMID: 37337103 PMCID: PMC10307629 DOI: 10.1038/s41590-023-01530-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/08/2023] [Indexed: 06/21/2023]
Abstract
Macrophages are involved in immune defense, organogenesis and tissue homeostasis. Macrophages contribute to the different phases of mammary gland remodeling during development, pregnancy and involution postlactation. Less is known about the dynamics of mammary gland macrophages in the lactation stage. Here, we describe a macrophage population present during lactation in mice. By multiparameter flow cytometry and single-cell RNA sequencing, we identified a lactation-induced CD11c+CX3CR1+Dectin-1+ macrophage population (liMac) that was distinct from the two resident F4/80hi and F4/80lo macrophage subsets present pregestationally. LiMacs were predominantly monocyte-derived and expanded by proliferation in situ concomitant with nursing. LiMacs developed independently of IL-34, but required CSF-1 signaling and were partly microbiota-dependent. Locally, they resided adjacent to the basal cells of the alveoli and extravasated into the milk. We found several macrophage subsets in human milk that resembled liMacs. Collectively, these findings reveal the emergence of unique macrophages in the mammary gland and milk during lactation.
Collapse
Affiliation(s)
- Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Roche, Basel, Switzerland
| | - Ekaterina Petrova
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Caroline Mussak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Christina A Welsh
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Wiebke Mildenberger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Ile-de-France, France
| | - Victor Kreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Elsa Roussel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian A Stifter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Myrto Andreadou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Camille Blériot
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut Necker des Enfants Malades, CNRS, Paris, France
| | - Francesca Ronchi
- University Clinic for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrew J Macpherson
- University Clinic for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Giancarlo Natalucci
- Larsson-Rosenquist Center for Neurodevelopment, Growth and Nutrition of the Newborn, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
- Newborn Research, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Schouten I, Bernys-Karolys A, Schneider P, Dror T, Ofer L, Shimoni C, Nissim-Eliraz E, Shpigel NY, Schlesinger S. Mesenchymal stromal cells modulate infection and inflammation in the uterus and mammary gland. BMC Vet Res 2023; 19:64. [PMID: 36997964 PMCID: PMC10061880 DOI: 10.1186/s12917-023-03616-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
The use of mesenchymal stromal cells (MSCs) is emerging as an efficacious and safe treatment for many infectious and non-infectious inflammatory diseases in human and veterinary medicine. Such use could be done to treat mastitis and metritis, which are the most common disease conditions affecting dairy cows leading to considerable economic losses and reduced animal welfare. Currently, both disease conditions are commonly treated using local and systemic administration of antibiotics. However, this strategy has many disadvantages including low cure rates and the public health hazards. Looking for alternative approaches, we investigated the properties of MSCs using in-vitro mammary and endometrial cell systems and in-vivo mastitis and metritis murine model systems. In-vitro, co-culture of mammary and uterus epithelial cells constructed with NF-kB reporter system, the master regulator of inflammation, demonstrated their anti-inflammatory effects in response to.LPS. In vivo, we challenge animals with field strains of mammary and utero pathogenic Escherichia coli and evaluated the effects of local and systemic application of MSC in the animal models. Disease outcome was evaluated using histological analysis, bacterial counts and gene expression of inflammatory markers. We show that MSC treatment reduced bacterial load in metritis and significantly modulated the inflammatory response of the uterus and mammary gland to bacterial infection. Most notably are the immune modulatory effects of remotely engrafted intravenous MSCs, which open new avenues to the development of MSC-based cell-free therapies.
Collapse
Affiliation(s)
- Iftach Schouten
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Andrés Bernys-Karolys
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Peleg Schneider
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Tal Dror
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Lior Ofer
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Chen Shimoni
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Einat Nissim-Eliraz
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel
| | - Nahum Y Shpigel
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel.
| | - Sharon Schlesinger
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, POB 12, Rehovot, 76100, Israel.
| |
Collapse
|
9
|
Rainard P, Gilbert FB, Germon P. Immune defenses of the mammary gland epithelium of dairy ruminants. Front Immunol 2022; 13:1031785. [PMID: 36341445 PMCID: PMC9634088 DOI: 10.3389/fimmu.2022.1031785] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The epithelium of the mammary gland (MG) fulfills three major functions: nutrition of progeny, transfer of immunity from mother to newborn, and its own defense against infection. The defense function of the epithelium requires the cooperation of mammary epithelial cells (MECs) with intraepithelial leucocytes, macrophages, DCs, and resident lymphocytes. The MG is characterized by the secretion of a large amount of a nutrient liquid in which certain bacteria can proliferate and reach a considerable bacterial load, which has conditioned how the udder reacts against bacterial invasions. This review presents how the mammary epithelium perceives bacteria, and how it responds to the main bacterial genera associated with mastitis. MECs are able to detect the presence of actively multiplying bacteria in the lumen of the gland: they express pattern recognition receptors (PRRs) that recognize microbe-associated molecular patterns (MAMPs) released by the growing bacteria. Interactions with intraepithelial leucocytes fine-tune MECs responses. Following the onset of inflammation, new interactions are established with lymphocytes and neutrophils recruited from the blood. The mammary epithelium also identifies and responds to antigens, which supposes an antigen-presenting capacity. Its responses can be manipulated with drugs, plant extracts, probiotics, and immune modifiers, in order to increase its defense capacities or reduce the damage related to inflammation. Numerous studies have established that the mammary epithelium is a genuine effector of both innate and adaptive immunity. However, knowledge gaps remain and newly available tools offer the prospect of exciting research to unravel and exploit the multiple capacities of this particular epithelium.
Collapse
|
10
|
Quiroga J, Vidal S, Siel D, Caruffo M, Valdés A, Cabrera G, Lapierre L, Sáenz L. Novel Proteoliposome-Based Vaccine against E. coli: A Potential New Tool for the Control of Bovine Mastitis. Animals (Basel) 2022; 12:ani12192533. [PMID: 36230275 PMCID: PMC9558995 DOI: 10.3390/ani12192533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Accepted: 09/18/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Mastitis is a highly prevalent disease in dairy cattle, affecting animal welfare and generating economic losses for the dairy industry. Control measures for coliform mastitis are limited, due to the constant exposure of the teat to bacteria and the emergence of antimicrobial-resistant bacteria, making vaccination an important strategy for control of mastitis. However, currently available vaccines show limited efficacy, which could be attributed to inactivation processes that alter the antigenic preservation of the vaccines. The aim of this study was to assess the efficacy of a novel vaccine against mastitis using proteoliposomes obtained from E. coli in a murine model of coliform mastitis. We demonstrated that the proteoliposome vaccine was safe, immunogenic and effective against an experimental model of E. coli mastitis, decreasing bacterial count and tissue damage. This proteoliposome vaccine is a potential new tool for prevention of mastitis. Abstract Escherichia coli is an important causative agent of clinical mastitis in cattle. Current available vaccines have shown limited protection. We evaluated the efficacy of a novel vaccine based on bacterial proteoliposomes derived from an E. coli field strain. Female BALB/c mice were immunized subcutaneously with two doses of the vaccine, 3 weeks apart. Between days 5 and 8 after the first inoculation, the females were mated. At 5–8 days postpartum, the mice were intramammary challenged with the same E. coli strain. Two days after bacterial infection, mice were euthanized, and the mammary glands were examined and removed to evaluate the efficacy and safety of the vaccine as well as the immune response generated by the new formulation. The vaccinated mice showed mild clinical symptoms and a lower mammary bacterial load as compared to non-vaccinated animals. The vaccination induced an increase in levels of IgG, IgG1 and IgG2a against E. coli in blood and mammary glands that showed less inflammatory infiltration and tissue damage, as compared to the control group. In summary, the vaccine based on bacterial proteoliposomes is safe, immunogenic, and effective against E. coli, constituting a new potential tool for mastitis control.
Collapse
Affiliation(s)
- John Quiroga
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Sonia Vidal
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
| | - Daniela Siel
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370035, Chile
| | - Mario Caruffo
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomás, Santiago 8370003, Chile
| | - Andrea Valdés
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
| | - Gonzalo Cabrera
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Lissette Lapierre
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
- Correspondence: (L.L.); (L.S.); Tel.: +56-9229-785689 (L.S.)
| | - Leonardo Sáenz
- Faculty of Veterinary Sciences, Universidad de Chile, Santiago 8820808, Chile
- Correspondence: (L.L.); (L.S.); Tel.: +56-9229-785689 (L.S.)
| |
Collapse
|
11
|
Alan A, Alan E, Arslan K, Daldaban F, Aksel EG, Çınar MU, Akyüz B. LPS- and LTA-Induced Expression of TLR4, MyD88, and TNF-α in Lymph Nodes of the Akkaraman and Romanov Lambs. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-15. [PMID: 36062368 DOI: 10.1017/s1431927622012314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Toll-like receptor (TLR)-mediated inflammatory processes play a critical role in the innate immune response during the initial interaction between the infecting microorganism and immune cells. This study aimed to investigate the possible microanatomical and histological differences in mandibular and bronchial lymph nodes in Akkaraman and Romanov lambs induced by lipopolysaccharide (LPS) and lipoteichoic acid (LTA) and study the gene, protein, and immunoexpression levels of TLR4, myeloid differentiation factor 88 (MyD88), and tumor necrosis factor-α (TNF-α) that are involved in the immune system. Microanatomical examinations demonstrated more intense lymphocyte infiltration in the bronchial lymph nodes of Akkaraman lambs in the LPS and LTA groups compared to Romanov lambs. TLR4, MyD88, and TNF-α immunoreactivities were more intense in the experimental groups of both breeds. Expression levels of MyD88 and TNF-α genes in the bronchial lymph node of Akkaraman lambs were found to increase statistically significantly in the LTA group. TLR4 gene expression level in the mandibular lymph node was found to be statistically significantly higher in the LTA + LPS group. In conclusion, dynamic changes in the immune cell populations involved in response to antigens such as LTA and LPS in the lymph nodes of both breeds can be associated with the difference in the expression level of the TLR4/MyD88/TNF-α genes.
Collapse
Affiliation(s)
- Aydın Alan
- Department of Anatomy, Faculty of Veterinary Medicine, Erciyes University, 38030 Kayseri, Turkey
| | - Emel Alan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, 38030 Kayseri, Turkey
| | - Korhan Arslan
- Department of Genetics, Faculty of Veterinary Medicine, University of Erciyes, 38030 Kayseri, Turkey
| | - Fadime Daldaban
- Department of Genetics, Faculty of Veterinary Medicine, University of Erciyes, 38030 Kayseri, Turkey
| | - Esma Gamze Aksel
- Department of Genetics, Faculty of Veterinary Medicine, University of Erciyes, 38030 Kayseri, Turkey
| | - Mehmet Ulaş Çınar
- Department of Animal Science, Faculty of Agriculture, University of Erciyes, 38030 Kayseri, Turkey
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA 99164, USA
| | - Bilal Akyüz
- Department of Genetics, Faculty of Veterinary Medicine, University of Erciyes, 38030 Kayseri, Turkey
| |
Collapse
|
12
|
Lv G, Wang H, Zhou X, Lian S, Wang J, Wu R. Effects of Hormone, NEFA and SCFA on the Migration of Neutrophils and the Formation of Neutrophil Extracellular Traps in Dairy Cows. Animals (Basel) 2022; 12:ani12091190. [PMID: 35565616 PMCID: PMC9103860 DOI: 10.3390/ani12091190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Perinatal dairy cows face the challenge of maintaining the resilience of defense against invading pathogens. During the perinatal period, hormonal or metabolic changes cause the decline of immune function of dairy cows and further lead to varying degrees of immunosuppression. The results of this study indicate that, hormones, nonesterified fatty acids (NEFAs) and short-chain fatty acids (SCFAs) can regulate neutrophil migration and the NETs formation of dairy cows in vitro. These results help to further explain the effects of changes in hormone secretion and metabolites on immunosuppression and the increased risk of disease in perinatal dairy cows. Abstract Polymorphonuclear neutrophils (PMN) are the first line of defense against the invasion of foreign pathogenic microorganisms and play an essential role in the immune system of dairy cows. The changes in hormone secretion and metabolites of dairy cows during the perinatal period are the key factors that cause immunosuppression and increased risk of diseases. However, the effects of the hormone, nonesterified fatty acid (NEFA), and short-chain fatty acid (SCFA) on the transmammary epithelial migration of dairy cows and the formation of neutrophil extracellular traps (NETs) have rarely been studied. This study explored the effects of hormones, NEFAs and SCFAs on the neutrophil migration and NETs formation of dairy cows in vitro. It was found that P4 and Ac can regulate the transepithelial migration of PMN; SA and Pr can regulate the formation of NETs; E2, OA and Bt can regulate PMN transepithelial migration and NET formation. These results help to further explain the effects of changes in hormone secretion and metabolites on immunosuppression and the increased risk of disease in perinatal dairy cows.
Collapse
Affiliation(s)
- Guanxin Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Hai Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Xiechen Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.L.); (H.W.); (X.Z.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
- Correspondence: ; Tel.: +86-459-6819188
| |
Collapse
|
13
|
Niikura M, Fukutomi T, Mineo S, Mitobe J, Kobayashi F. Malaria in the postpartum period causes damage to the mammary gland. PLoS One 2021; 16:e0258491. [PMID: 34644348 PMCID: PMC8513860 DOI: 10.1371/journal.pone.0258491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
Mastitis is an inflammation of the mammary gland in the breast and is typically due to bacterial infection. In malaria-endemic areas, mastitis with accompanying fever can be challenging to differentiate from malaria. At the same time, it is unclear whether malaria infection is directly involved in the development of mastitis. In the present study, whether mastitis develops during infection with malaria parasites was investigated using a rodent malaria model with Plasmodium berghei (P. berghei; Pb) ANKA. The course of parasitemia in postpartum mice infected with Pb ANKA was similar to the course in infected virgin mice. However, infected postpartum mice died earlier than did infected virgin mice. In addition, the weight of pups from mice infected with Pb ANKA was significantly reduced compared with pups from uninfected mice. The macroscopic and histological analyses showed apparent changes, such as destruction of the alveolus wall and extensive presence of leukocytes, in mammary gland tissue in mice infected during the postpartum period. The findings suggest that women during the postpartum period are more vulnerable to complications when infected with malaria parasites, particularly women who do not acquire protective immunity against malaria parasites. Based on the proteomic analysis, IFN-γ signaling pathway-related proteins in mammary gland tissue of the infected postpartum mice were increased. Our results indicate that inflammation induced by IFN-γ, a proinflammatory cytokine, may contribute to negative histological changes in mammary gland tissue of postpartum mice infected with Pb ANKA. In IFN-γ receptor 1-deficient (IFNGR1-KO) mice, the histological changes in mammary gland tissue of the infected postpartum wild-type mice were improved to almost normal mammary gland structure. Furthermore, weight loss in pups delivered by infected IFNGR1-KO postpartum mice was not observed. Taken together, these findings indicate that inflammation induced by IFN-γ is associated with development of mastitis in postpartum mice infected with Pb ANKA. The present study results may increase our understanding of how disease aggravation occurs during postpartum malaria.
Collapse
Affiliation(s)
- Mamoru Niikura
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| | - Toshiyuki Fukutomi
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo, Japan
| | - Shoichiro Mineo
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Jiro Mitobe
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| | - Fumie Kobayashi
- Department of Environmental Science, School of Life and Environmental Science, Azabu University, Kanagawa, Japan
| |
Collapse
|
14
|
Regulation of ydiV-induced biological characteristics permits Escherichia coli evasion of the host STING inflammatory response. Vet Microbiol 2021; 261:109207. [PMID: 34419774 DOI: 10.1016/j.vetmic.2021.109207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022]
Abstract
Mammary gland-derived Escherichia coli (E. coli) is an important pathogen causing dairy cow mastitis. YdiV, with EAL-like domains, inhibits flagellum biogenesis and motility and affects c-di-GMP (eubacterial signaling molecule) concentration changes in bacteria. However, the pathophysiological role of ydiV in host-pathogen cross-talk still needs to be elucidated. In this study, firstly constructed the ydiV mutant (NJ17ΔydiV) and ydiV complementary (cNJ17ΔydiV) E. coli strains to infect mouse mammary epithelial cells (EpH4-Ev) and macrophages (RAW264.7), as well as mouse mammary glands, respectively. Then biological characteristics, adaptor molecules in related signaling pathways, proinflammatory cytokines and the extent of host cell damage was evaluated. Compared with E. coli NJ17 infected mice, the bacterial load in the mammary gland of NJ17ΔydiV was significantly lower and the extent of the damage was alleviated. Notably, the deletion of ydiV significantly aggravated cell damage in RAW264.7 cells and compared with the wild-type strain, NJ17ΔydiV significantly activated the STING/TBK1/IRF3 pathway in macrophages. In EpH4-Ev cells, although STING did not sense E. coli NJ17 invasion, IRF3 was activated by the NJ17ΔydiV strain. Taken together, ydiV deletion significantly affects a variety of biological characteristics and induces severe cell damage, while the STING/TBK1/IRF3 pathway actively participated in pathogen elimination in the host. This study highlights a new role for ydiV in E. coli infection and provides a foundation for further studies to better understand host-bacteria interactions and potential prophylactic strategies for infectious diseases.
Collapse
|
15
|
The deletion of yeaJ gene facilitates Escherichia coli escape from immune recognition. J Bacteriol 2021; 203:e0033621. [PMID: 34309400 DOI: 10.1128/jb.00336-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mammary gland-derived Escherichia coli (E. coli) is an important pathogen causing dairy cow mastitis. Mammary gland mucosal immunity against infectious E. coli mainly depends on recognition of pathogen-associated molecular patterns by innate receptors. Stimulator of interferon (IFN) gene (STING) has recently been the dominant mediator in reacting to bacterial intrusion and preventing inflammatory disorders. In this study, we firstly proved that diguanylate cyclase YeaJ relieves mouse mammary gland pathological damage by changing E. coli phenotypic and host STING-dependent innate immunity response. YeaJ decreases mammary gland circular vacuoles, bleeding and degeneration in mice. In addition, YeaJ participates in STING-IRF3 signaling to regulate inflammation in vivo. While in vitro, YeaJ decreases damage to macrophages (RAW264.7) but not to mouse mammary epithelial cells (EpH4-Ev). Consistent with the results in mouse mammary gland, yeaJ significantly activates STING/TBK1/IRF3 pathway in RAW264.7 as well. In conclusion, the deletion of yeaJ gene facilitates E. coli NJ17 escape from STING-dependent innate immunity recognition in vitro and in vivo. This study highlights a novel role for YeaJ in E. coli infection, which provides a better understanding of host-bacteria interactions and potential prophylactic strategies for infections. IMPORTANCE E. coli is the etiological agent of environmental mastitis in dairy cows, which cause massive financial losses worldwide. However, the pathophysiological role of yeaJ in the interaction between E. coli and host remains unclear. We found that YeaJ significantly influences various biological characteristics and suppresses severe inflammatory response as well as greater damage. YeaJ alleviates damage to macrophages (RAW264.7) and mouse mammary gland. Moreover, these effects of YeaJ are achieved at least partial by mediating the STING-IRF3 signaling pathway. In conclusion, the deletion of yeaJ gene facilitates E. coli NJ17 escape from STING-dependent innate immunity recognition in vitro and in vivo. This study is the basis for further research to better understand host-bacteria interactions and provides potential prophylactic strategies for infections.
Collapse
|
16
|
Ge BJ, Zhao P, Li HT, Sang R, Wang M, Zhou HY, Zhang XM. Taraxacum mongolicum protects against Staphylococcus aureus-infected mastitis by exerting anti-inflammatory role via TLR2-NF-κB/MAPKs pathways in mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113595. [PMID: 33212175 DOI: 10.1016/j.jep.2020.113595] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/06/2020] [Accepted: 11/12/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine, Taraxacum mongolicum has been widely used for the prevention and treatment of a variety of inflammatory and infectious diseases, and also clinically used as a remedy for mastitis. However, the scientific rationale and mechanism behind its use on mastitis in vivo are still unclear. AIM OF THE STUDY This study aimed to investigate the protective effect and potential mechanism of Taraxacum mongolicum Hand.-Mazz. (T. mongolicum) on mastitis infected by Staphylococcus aureus (S. aureus). MATERIALS AND METHODS Female ICR mice were given intragastrically 2.5, 5 and 10 g/kg of T. mongolicum extract twice per day for 6 consecutive days, and infected with S. aureus via teat canal to induce mastitis. Pro-inflammatory cytokine tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) levels were determined by ELISA. Myeloperoxidase (MPO) activity and distribution were measured by reagent kit and immunohistochemistry. Histopathological changes of mammary gland tissues were observed by H&E staining. Toll-like receptor 2 (TLR2) expression, phosphorylations of related proteins in nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways were detected by western blot. RESULTS T. mongolicum decreased TNF-α, IL-6 and IL-1β levels, and reduced MPO activity and distribution in sera and mammary glands with S. aureus-infected mastitis. In addition, T. mongolicum effectively attenuated histopathological damages and cell necrosis of mammary gland tissues infected by S. aureus. Moreover, T. mongolicum inhibited the expression of TLR2, and the phosphorylations of inhibitor κBα (IκBα), p65, p38, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) proteins in mammary glands with S. aureus-infected mastitis. CONCLUSIONS This study suggests that T. mongolicum protects against S. aureus-infected mastitis by exerting anti-inflammatory role, which is attributed to the inhibition of TLR2-NF-κB/MAPKs pathways.
Collapse
Affiliation(s)
- Bing-Jie Ge
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| | - Peng Zhao
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| | - Hai-Tao Li
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China; Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Juye Street, Changchun, Jilin 132109, China.
| | - Rui Sang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| | - Meng Wang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| | - Hong-Yuan Zhou
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| | - Xue-Mei Zhang
- Agricultural College of Yanbian University, Gongyuan Street, Yanji, Jilin, 133002, China.
| |
Collapse
|
17
|
Salamon H, Nissim-Eliraz E, Ardronai O, Nissan I, Shpigel NY. The role of O-polysaccharide chain and complement resistance of Escherichia coli in mammary virulence. Vet Res 2020; 51:77. [PMID: 32539761 PMCID: PMC7294653 DOI: 10.1186/s13567-020-00804-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022] Open
Abstract
Mastitis, inflammation of the mammary gland, is a common disease of dairy animals. The disease is caused by bacterial infection ascending through the teat canal and mammary pathogenic Escherichia coli (MPEC) are common etiology. In the first phase of infection, virulence mechanisms, designated as niche factors, enable MPEC bacteria to resist innate antimicrobial mechanisms, replicate in milk, and to colonize the mammary gland. Next, massive replication of colonizing bacteria culminates in a large biomass of microbe-associated molecular patterns (MAMPs) recognized by pattern recognition receptors (PRRs) such as toll-like receptors (TLRs) mediating inflammatory signaling in mammary alveolar epithelial cells (MAEs) and macrophages. Bacterial lipopolysaccharides (LPSs), the prototypical class of MAMPs are sufficient to elicit mammary inflammation mediated by TLR4 signaling and activation of nuclear factor kB (NF-kB), the master regulator of inflammation. Using in vivo mastitis model, in low and high complements mice, and in vitro NF-kB luminescence reporter system in MAEs, we have found that the smooth configuration of LPS O-polysaccharides in MPEC enables the colonizing organisms to evade the host immune response by reducing inflammatory response and conferring resistance to complement. Screening a collection of MPEC field strains, we also found that all strains were complement resistant and 94% (45/48) were smooth. These results indicate that the structure of LPS O-polysaccharides chain is important for the pathogenesis of MPEC mastitis and provides protection against complement-mediated killing. Furthermore, we demonstrate a role for complement, a key component of innate immunity, in host-microbe interactions of the mammary gland.
Collapse
Affiliation(s)
- Hagit Salamon
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, POB 12, 76100, Rehovot, Israel
| | - Einat Nissim-Eliraz
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, POB 12, 76100, Rehovot, Israel
| | - Oded Ardronai
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, POB 12, 76100, Rehovot, Israel
| | - Israel Nissan
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, POB 12, 76100, Rehovot, Israel
| | - Nahum Y Shpigel
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, POB 12, 76100, Rehovot, Israel.
| |
Collapse
|
18
|
Penadés M, Viana D, García-Quirós A, Muñoz-Silvestre A, Moreno-Grua E, Pérez-Fuentes S, Pascual JJ, Corpa JM, Selva L. Differences in virulence between the two more prevalent Staphylococcus aureus clonal complexes in rabbitries (CC121 and CC96) using an experimental model of mammary gland infection. Vet Res 2020; 51:11. [PMID: 32054530 PMCID: PMC7020377 DOI: 10.1186/s13567-020-0740-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/19/2020] [Indexed: 01/03/2023] Open
Abstract
Staphylococcal mastitis is a major health problem in humans and livestock that leads to economic loss running in millions. This process is currently one of the main reasons for culling adult rabbit does. Surprisingly, the two most prevalent S. aureus lineages isolated from non-differentiable natural clinical mastitis in rabbits (ST121 and ST96) generate different immune responses. This study aimed to genetically compare both types of strains to search for possible dissimilarities to explain differences in immune response, and to check whether they showed similar virulence in in vitro tests as in experimental intramammary in vivo infection. The main differences were observed in the enterotoxin gene cluster (egc) and the immune-evasion-cluster (IEC) genes. While isolate ST121 harboured all six egc cluster members (seg, sei, selm, seln, selo, selu), isolate ST96 lacked the egc cluster. Strain ST96 carried a phage integrase Sa3 (Sa3int), compatible with a phage integrated into the hlb gene (β-haemolysin-converting bacteriophages) with IEC type F, while isolate ST121 lacked IEC genes and the hlb gene was intact. Moreover, the in vitro tests confirmed a different virulence capacity between strains as ST121 showed greater cytotoxicity for erythrocytes, polymorphonuclear leukocytes and macrophages than strain ST96. Differences were also found 7 days after experimental intramammary infection with 100 colony-forming units. The animals inoculated with strain ST121 developed more severe gross and histological mastitis, higher counts of macrophages in tissue and of all the cell populations in peripheral blood, and a significantly larger total number of bacteria than those infected by strain ST96.
Collapse
Affiliation(s)
- Mariola Penadés
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - David Viana
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - Ana García-Quirós
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - Asunción Muñoz-Silvestre
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - Elena Moreno-Grua
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - Sara Pérez-Fuentes
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain
| | - Juan José Pascual
- Institute for Animal Science and Technology, Universitat Politècnica de València, Camino de Vera 14, 46071, Valencia, Spain
| | - Juan M Corpa
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain.
| | - Laura Selva
- Biomedical Research Institute (PASAPTA-Pathology Group), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. C/Tirant lo Blanc 7, Alfara del Patriarca, 46115, Valencia, Spain.
| |
Collapse
|
19
|
Luan W, Liu X, Wang X, An Y, Wang Y, Wang C, Shen K, Xu H, Li S, Liu M, Yu LU. Inhibition of Drug Resistance of Staphylococcus aureus by Efflux Pump Inhibitor and Autolysis Inducer to Strengthen the Antibacterial Activity of β-lactam Drugs. Pol J Microbiol 2019; 68:477-491. [PMID: 31880892 PMCID: PMC7260704 DOI: 10.33073/pjm-2019-047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 01/17/2023] Open
Abstract
This study explored a potential treatment against methicillin-resistant Staphylococcus aureus (MRSA) infections that combines thioridazine (TZ), an efflux pump inhibitor, and miconazole (MCZ), an autolysis inducer, with the anti-microbial drug cloxacillin (CXN). In vitro, the combination treatment of TZ and MCZ significantly reduced 4096-fold (Σ (FIC) = 0.1 – 1.25) the MIC value of CXN against S. aureus. In vivo, the combination therapy significantly relieved breast redness and swelling in mice infected with either clinical or standard strains of S. aureus. Meanwhile, the number of bacteria isolated from the MRSA135-infected mice decreased significantly (p = 0.0427 < 0.05) after the combination therapy when compared to monotherapy. Moreover, the number of bacteria isolated from the mice infected with a reference S. aureus strain also decreased significantly (p = 0.0191 < 0.05) after the combination therapy when compared to monotherapy. The pathological changes were more significant in the CXN-treated group when compared to mice treated with a combination of three drugs. In addition, we found that combination therapy reduced the release of the bacteria-stimulated cytokines such as IL-6, IFN-γ, and TNF-α. Cytokine assays in serum revealed that CXN alone induced IL-6, IFN-γ, and TNF-α in the mouse groups infected with ATCC 29213 or MRSA135, and the combination of these three drugs significantly reduced IL-6, IFN-γ, and TNF-α concentrations. Also, the levels of TNF-α and IFN-γ in mice treated with a combination of three drugs were significantly lower than in the CXN-treated group. Given the synergistic antibacterial activity of CXN, we concluded that the combination of CXN with TZ, and MCZ could be developed as a novel therapeutic strategy against S. aureus.
Collapse
Affiliation(s)
- Wenjing Luan
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Xiaolei Liu
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Xuefei Wang
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Yanan An
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Yang Wang
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Chao Wang
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Keshu Shen
- Jilin Hepatobiliary Hospital , Changchun , China
| | - Hongyue Xu
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Shulin Li
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| | - Mingyuan Liu
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - L U Yu
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Department of Infectious Diseases of First Hospital of Jilin University, College of Veterinary Medicine Jilin University , Changchun , China
| |
Collapse
|
20
|
Yadav RK, Tripathi CB, Saraf SA, Ansari MN, Saeedan AS, Aldosary S, Rajinikanth PS, Kaithwas G. Alpha-linolenic acid based nano-suspension protect against lipopolysaccharides induced mastitis by inhibiting NFκBp65, HIF-1α, and mitochondria-mediated apoptotic pathway in albino Wistar rats. Toxicol Appl Pharmacol 2019; 377:114628. [DOI: 10.1016/j.taap.2019.114628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/30/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
|
21
|
Hu X, Zhang N, Fu Y. Role of Liver X Receptor in Mastitis Therapy and Regulation of Milk Fat Synthesis. J Mammary Gland Biol Neoplasia 2019; 24:73-83. [PMID: 30066175 DOI: 10.1007/s10911-018-9403-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/04/2018] [Indexed: 02/03/2023] Open
Abstract
Mastitis is important disease that causes huge economic losses in the dairy industry. In recent years, antibiotic therapy has become the primary treatment for mastitis, however, due to drug residue in milk and food safety factors, we lack safe and effective drugs for treating mastitis. Therefore, new targets and drugs are urgently needed to control mastitis. LXRα, one of the main members of the nuclear receptor superfamily, is reported to play important roles in metabolism, infection and immunity. Activation of LXRα could inhibit LPS-induced mastitis. Furthermore, LXRα is reported to enhance milk fat production, thus, LXRα may serve as a new target for mastitis therapy and regulation of milk fat synthesis. This review summarizes the effects of LXRα in regulating milk fat synthesis and treatment of mastitis and highlights the potential agonists involved in both issues.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Cattle
- Dairying
- Escherichia coli/isolation & purification
- Escherichia coli/pathogenicity
- Female
- Global Burden of Disease
- Humans
- Immunity, Innate
- Lactation/metabolism
- Lipid Metabolism
- Liver X Receptors/agonists
- Liver X Receptors/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/microbiology
- Mammary Glands, Animal/pathology
- Mammary Glands, Human/cytology
- Mammary Glands, Human/immunology
- Mammary Glands, Human/microbiology
- Mammary Glands, Human/pathology
- Mastitis/drug therapy
- Mastitis/immunology
- Mastitis/microbiology
- Mastitis, Bovine/drug therapy
- Mastitis, Bovine/epidemiology
- Mastitis, Bovine/immunology
- Mastitis, Bovine/microbiology
- Membrane Microdomains/metabolism
- Milk/metabolism
- Prevalence
- Receptors, Pattern Recognition/metabolism
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, People's Republic of China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, People's Republic of China.
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, People's Republic of China.
| |
Collapse
|
22
|
IL-1β directly inhibits milk lipid production in lactating mammary epithelial cells concurrently with enlargement of cytoplasmic lipid droplets. Exp Cell Res 2018; 370:365-372. [DOI: 10.1016/j.yexcr.2018.06.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
|
23
|
Breyne K, Steenbrugge J, Demeyere K, Lee CG, Elias JA, Petzl W, Smith DGE, Germon P, Meyer E. Immunomodulation of Host Chitinase 3-Like 1 During a Mammary Pathogenic Escherichia coli Infection. Front Immunol 2018; 9:1143. [PMID: 29892291 PMCID: PMC5985307 DOI: 10.3389/fimmu.2018.01143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/07/2018] [Indexed: 12/05/2022] Open
Abstract
Chitin is a N-acetyl-d-glucosamine biopolymer that can be recognized by chitin-binding proteins. Although mammals lack chitin synthase, they induce proteins responsible for detecting chitin in response to bacterial infections. Our aim was to investigate whether chitinase 3-like 1 (CHI3L1) has a potential role in the innate immunity of the Escherichia coli (E. coli) infected mammary gland. CHI3L1 protein was found to be secreted in whey of naturally coliform-affected quarters compared to whey samples isolated from healthy udders. In addition, gene expression of CHI3L1 was confirmed in udder tissue of cows experimentally infected with a mammary pathogenic E. coli (MPEC) strain. Despite the known anatomical differences, the bovine udders’ innate immune response was mimicked by applying an experimental mouse model using MPEC or non-MPEC isolates. The effect of CHI3L1 expression in the murine mammary gland in response to coliform bacteria was investigated through the use of CHI3L1−/− mice as well as through treatment with either a pan-caspase inhibitor or chitin particles in wild-type mice. The local induction of CHI3L1 postinfection with different E. coli strains was demonstrated to be independent of both bacterial growth and mammary interleukin (IL)-8 levels. Indeed, CHI3L1 emerged as a regulator impacting on the transcytosis of Ly6G-positive cells from the interstitial space into the alveolar lumen of the mammary tissue. Furthermore, CHI3L1 was found to be upstream regulated by caspase activity and had a major downstream effect on the local pro-inflammatory cytokine profile, including IL-1beta, IL-6, and RANTES/CCL5. In conclusion, CHI3L1 was demonstrated to play a key role in the cytokine and caspase signaling during E. coli triggered inflammation of the mammary gland.
Collapse
Affiliation(s)
- Koen Breyne
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chun Geun Lee
- Division of Biology and Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, United States
| | - Jack A Elias
- Division of Biology and Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, United States
| | - Wolfram Petzl
- Clinic for Ruminants with Ambulance and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - David G E Smith
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
| | - Pierre Germon
- INRA UMR 1282 Infectiologie et Santé Publique (ISP), Université François Rabelais de Tours, Nouzilly, France
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
24
|
Luteolin reduces inflammation in Staphylococcus aureus-induced mastitis by inhibiting NF-kB activation and MMPs expression. Oncotarget 2018; 8:28481-28493. [PMID: 28415707 PMCID: PMC5438666 DOI: 10.18632/oncotarget.16092] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 11/29/2022] Open
Abstract
Mastitis is a serious and prevalent disease caused by infection by pathogens such as Staphylococcus aureus. We evaluated the anti-inflammatory effects and mechanism of luteolin, a natural flavonoid with a wide range of pharmacological activities, in a mouse model of S. aureus mastitis. We also treated cultured mouse mammary epithelial cells (mMECs) with S. aureus and luteolin. Histopathological changes were examined by H&E staining and the levels of inflammatory cytokine proteins were analyzed using ELISAs. We determined mRNA levels with qPCR and the level of NF-κB and matrix metalloproteinase (MMP) proteins by Western blotting. The observed histopathological changes showed that luteolin protected mammary glands with S. aureus infection from tissue destruction and inflammatory cell infiltration. Luteolin inhibited the expression of TNF-α, IL-1β, and IL-6, all of which were increased with S. aureus infection of mammary tissues and mMECs. S. aureus-induced TLR2 and TLR4 was suppressed by luteolin, as were levels of IκBα and NF-κB p65 phosphorylation and expression of MMP-2 and MMP-9. Levels of tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 were enhanced. These findings suggest luteolin is a potentially effective new treatment to reduce tissue damage and inflammation from S. aureus-induced mastitis.
Collapse
|
25
|
Petzl W, Zerbe H, Günther J, Seyfert HM, Hussen J, Schuberth HJ. Pathogen-specific responses in the bovine udder. Models and immunoprophylactic concepts. Res Vet Sci 2017; 116:55-61. [PMID: 29275905 DOI: 10.1016/j.rvsc.2017.12.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/28/2017] [Accepted: 12/17/2017] [Indexed: 12/20/2022]
Abstract
Bovine mastitis is a disease of major economic effects on the dairy industry worldwide. Experimental in vivo infection models have been widely proven as an effective tool for the investigation of pathogen-specific host immune responses. Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) are two common mastitis pathogens with an opposite clinical outcome of the disease. E. coli and S. aureus have proven to be valid surrogates to model clinical and subclinical mastitis respectively. Contemporary transcriptome profiling studies demonstrated that the transcriptomic response in the teat reflects the course of pathogen-specific mastitis, being ultimately determined by the immune response of the mammary epithelial cells. After an experimental in vivo challenge, E. coli induces a vigorous early transcriptional response in udder tissue being quantitatively and - notably - qualitatively distinct from the much weaker response against an S. aureus infection. E. coli mastitis models proved that the local response in the infected udder quarters is accompanied by a response in non-infected neighbouring udder quarters modulating systemically their immune responsiveness. Immunomodulation of the udder was investigated in animal models. Pathophysiological consequences were studied after intramammary administration of cytokines, chemokines, growth factors, steroidal anti-inflammatory drugs, or priming of tissue resident cells with pathogen-derived molecules. The latter approaches resulted only in a temporal protection of the udder, reducing transiently the risk of infection but sustained lowering of the severity of an eventually occurring mastitis. They offer an alternative to vaccination trials, which over decades also did not yield protection against new infections.
Collapse
Affiliation(s)
- Wolfram Petzl
- Clinic for Ruminants with Ambulance and Herd Health Services, Centre for Clinical Veterinary Medicine, LMU Munich, Germany
| | - Holm Zerbe
- Clinic for Ruminants with Ambulance and Herd Health Services, Centre for Clinical Veterinary Medicine, LMU Munich, Germany
| | - Juliane Günther
- Leibniz Institute for Farm Animal Biology, Institute for Genome Biology, Dummerstorf, Germany
| | - Hans-Martin Seyfert
- Leibniz Institute for Farm Animal Biology, Institute for Genome Biology, Dummerstorf, Germany
| | - Jamal Hussen
- Department of Microbiology and Parasitology, College of Veterinary Medicine, King Faisal University, Al Ahsaa, Saudi Arabia
| | | |
Collapse
|
26
|
Xiao HB, Sui GG, Lu XY, Sun ZL. Kaempferol modulates Angiopoietin-like protein 2 expression to lessen the mastitis in mice. Pharmacol Rep 2017; 70:439-445. [PMID: 29627690 DOI: 10.1016/j.pharep.2017.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 11/05/2017] [Accepted: 11/20/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mastitis is inflammation of a breast (or udder). Angiopoietin-like protein 2 (ANGPTL2) has been found as a key inflammatory mediator in mastitis. Purpose of this research was to investigate the mechanisms about repressing effect of kaempferol on mastitis. METHODS Forty mice were randomly divided into 4 groups (n=10): C57BL/6J control mice, untreated murine mastitis, 10mg/kg kaempferol treated murine mastitis (ip), and 30mg/kg kaempferol treated murine mastitis (ip). Primary cultured mouse mammary epithelial cells (MMEC) were indiscriminately divided into seven groups including control group, 10mmol/L vehicle of kaempferol group, 10μmol/L kaempferol treated group, 20μg/mL LPS treated group, 1μmol/L kaempferol plus LPS treated group, 3μmol/L kaempferol plus LPS treated group, and 10μmol/L kaempferol plus LPS treated group. RESULTS In murine mastitis, kaempferol (10 or 30mg/kg) treatment prevented mastitis development, decreased myeloperoxidase (MPO) production, interleukin (IL)-6 level, tumour necrosis factor-α (TNF-α) concentration, and ANGPTL2 expression. In MMEC, kaempferol (1, 3 or 10μM) reduced MPO production, TNF-α concentration, IL-6 level, and ANGPTL2 expression. CONCLUSIONS The results in present study show that kaempferol modulates the expression of ANGPTL2 to lessen the mastitis in mice.
Collapse
Affiliation(s)
- Hong-Bo Xiao
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| | - Guo-Guang Sui
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xiang-Yang Lu
- Hunan Province University Key Laboratory for Agricultural Biochemistry and Biotransformation, Hunan Agricultural University, Changsha, China; Hunan Co-Innovation Center for Ultilization of Botanical Functional Ingredients, Changsha, China
| | - Zhi-Liang Sun
- Biological Veterinary Drugs Branch, National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, China
| |
Collapse
|
27
|
Elevated Levels of ADMA Are Associated with Lower DDAH2 and Higher PRMT1 in LPS-Induced Endometritis Rats. Inflammation 2017; 41:299-306. [DOI: 10.1007/s10753-017-0687-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
28
|
In vivo imaging of lung inflammation with neutrophil-specific 68Ga nano-radiotracer. Sci Rep 2017; 7:13242. [PMID: 29038592 PMCID: PMC5643527 DOI: 10.1038/s41598-017-12829-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/15/2017] [Indexed: 02/05/2023] Open
Abstract
In vivo detection and quantification of inflammation is a major goal in molecular imaging. Furthermore, cell-specific detection of inflammation would be a tremendous advantage in the characterization of many diseases. Here, we show how this goal can be achieved through the synergistic combination of nanotechnology and nuclear imaging. One of the most remarkable features of this hybrid approach is the possibility to tailor the pharmacokinetics of the nanomaterial-incorporated biomolecule and radionuclide. A good example of this approach is the covalent binding of a large amount of a neutrophil-specific, hydrophobic peptide on the surface of 68Ga core-doped nanoparticles. This new nano-radiotracer has been used for non-invasive in vivo detection of acute inflammation with very high in vivo labelling efficiency, i.e. a large percentage of labelled neutrophils. Furthermore, we demonstrate that the tracer is neutrophil-specific and yields images of neutrophil recruitment of unprecedented quality. Finally, the nano-radiotracer was successfully detected in chronic inflammation in atherosclerosis-prone ApoE−/− mice after several weeks on a high-fat diet.
Collapse
|
29
|
Breyne K, Steenbrugge J, Demeyere K, Vanden Berghe T, Meyer E. Preconditioning with Lipopolysaccharide or Lipoteichoic Acid Protects against Staphylococcus aureus Mammary Infection in Mice. Front Immunol 2017; 8:833. [PMID: 28791009 PMCID: PMC5522847 DOI: 10.3389/fimmu.2017.00833] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is one of the most causative agents of mastitis and is associated with chronic udder infections. The persistency of the pathogen is believed to be the result of an insufficient triggering of local inflammatory signaling. In this study, the preclinical mastitis model was used, aiming to evaluate if lipopolysaccharide (LPS) or lipoteichoic acid (LTA) preconditioning could aid the host in more effectively clearing or at least limiting a subsequent S. aureus infection. A prototypic Gram-negative virulence factor, i.e., LPS and Gram-positive virulence factor, i.e., LTA were screened whether they were able to boost the local immune compartment. Compared to S. aureus-induced inflammation, both toxins had a remarkable high potency to efficiently induce two novel selected innate immunity biomarkers i.e., lipocalin 2 (LCN2) and chitinase 3-like 1 (CHI3L1). When combining mammary inoculation of LPS or LTA prior to a local S. aureus infection, we were able to modulate the innate immune response, reduce local bacterial loads, and induce either LCN2 or CHI3L1 at 24 h post-infection. Clodronate depletion of mammary macrophages also identified that macrophages contribute only to a limited extend to the LPS/LTA-induced immunomodulation upon S. aureus infection. Based on histological neutrophil influx evaluation, concomitant local cytokine profiles and LCN2/CHI3L1 patterns, the macrophage-independent signaling plays a major role in the LPS- or LTA-pretreated S. aureus-infected mouse mammary gland. Our results highlight the importance of a vigilant microenvironment during the innate immune response of the mammary gland and offer novel insights for new approaches concerning effective immunomodulation against a local bacterial infection.
Collapse
Affiliation(s)
- Koen Breyne
- Biochemistry, Faculty of Veterinary Medicine, Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| | - Jonas Steenbrugge
- Biochemistry, Faculty of Veterinary Medicine, Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Biochemistry, Faculty of Veterinary Medicine, Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| | - Tom Vanden Berghe
- Peter Vandenabeele Lab, Inflammation Research Center, Department of Biomedical Molecular Biology, VIB, Ghent University, Zwijnaarde, Belgium
| | - Evelyne Meyer
- Biochemistry, Faculty of Veterinary Medicine, Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| |
Collapse
|
30
|
Roussel P, Porcherie A, Répérant-Ferter M, Cunha P, Gitton C, Rainard P, Germon P. Escherichia coli mastitis strains: In vitro phenotypes and severity of infection in vivo. PLoS One 2017; 12:e0178285. [PMID: 28727781 PMCID: PMC5519002 DOI: 10.1371/journal.pone.0178285] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/10/2017] [Indexed: 01/18/2023] Open
Abstract
Mastitis remains a major infection of dairy cows and an important issue for dairy farmers and the dairy industry, in particular infections due to Escherichia coli strains. So far, properties specific to E. coli causing mastitis remain ill defined. In an attempt to better understand the properties required for E. coli to trigger mastitis, we used a range of in vitro assays to phenotypically characterize four E. coli strains, including the prototypical E. coli mastitis strain P4, possessing different relative abilities to cause mastitis in a mouse model. Our results indicate that a certain level of serum resistance might be required for colonization of the mammary gland. Resistance to neutrophil killing is also likely to contribute to a slower clearance of bacteria and higher chances to colonize the udder. In addition, we show that the four different strains do induce a pro-inflammatory response by mammary epithelial cells but with different intensities. Interestingly, the prototypical mastitis strain P4 actually induces the less intense response while it is responsible for the most severe infections in vivo. Altogether, our results suggest that different strategies can be used by E. coli strains to colonize the mammary gland and cause mastitis.
Collapse
Affiliation(s)
- Perrine Roussel
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Adeline Porcherie
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | | | - Patricia Cunha
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Christophe Gitton
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Pascal Rainard
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Pierre Germon
- ISP, INRA, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
- * E-mail:
| |
Collapse
|
31
|
Detilleux J. A strategy to estimate the rate of recruitment of inflammatory cells during bovine intramammary infection under field management. BMC Vet Res 2017; 13:167. [PMID: 28595581 PMCID: PMC5465529 DOI: 10.1186/s12917-017-1078-4] [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: 09/27/2016] [Accepted: 05/26/2017] [Indexed: 11/10/2022] Open
Abstract
Background In most infectious diseases, among which bovine mastitis, promptness of the recruitment of inflammatory cells (mainly neutrophils) in inflamed tissues has been shown to be of prime importance in the resolution of the infection. Although this information should aid in designing efficient control strategies, it has never been quantified in field studies. Methods Here, a system of ordinary differential equations is proposed that describes the dynamic process of the inflammatory response to mammary pathogens. The system was tested, by principal differential analysis, on 1947 test-day somatic cell counts collected on 756 infected cows, from 50 days before to 50 days after the diagnosis of clinical mastitis. Cell counts were log-transformed before estimating recruitment rates. Results Daily rates of cellular recruitment was estimated at 0.052 (st. err. = 0.005) during health. During disease, an additional cellular rate of recruitment was estimated at 0.004 (st. err. = 0.001) per day and per bacteria. These estimates are in agreement with analogous measurements of in vitro neutrophil functions. Conclusions Results suggest the method is adequate to estimate one of the components of innate resistance to mammary pathogens at the individual level and in field studies. Extension of the method to estimate components of innate tolerance and limits of the study are discussed. Electronic supplementary material The online version of this article (doi:10.1186/s12917-017-1078-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J Detilleux
- Fundamental and Applied Research for Animals and Healh, Sustainable Animal Production, Faculty of Veterinary Medicine, University of Liège, Quartier Vallée 2, Avenue de Cureghem, 6, 4000, Liège, Belgium.
| |
Collapse
|
32
|
Estrogen reprograms the activity of neutrophils to foster protumoral microenvironment during mammary involution. Sci Rep 2017; 7:46485. [PMID: 28429725 PMCID: PMC5399373 DOI: 10.1038/srep46485] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/21/2017] [Indexed: 12/14/2022] Open
Abstract
Epidemiological studies have indicated increased risk for breast cancer within 10 years of childbirth. Acute inflammation during mammary involution has been suggested to promote this parity-associated breast cancer. We report here that estrogen exacerbates mammary inflammation during involution. Microarray analysis shows that estrogen induces an extensive proinflammatory gene signature in the involuting mammary tissue. This is associated with estrogen-induced neutrophil infiltration. Furthermore, estrogen induces the expression of protumoral cytokines/chemokines, COX-2 and tissue-remodeling enzymes in isolated mammary neutrophils and systemic neutrophil depletion abolished estrogen-induced expression of these genes in mammary tissue. More interestingly, neutrophil depletion diminished estrogen-induced growth of ERα-negative mammary tumor 4T1 in Balb/c mice. These findings highlight a novel aspect of estrogen action that reprograms the activity of neutrophils to create a pro-tumoral microenvironment during mammary involution. This effect on the microenvironment would conceivably aggravate its known neoplastic effect on mammary epithelial cells.
Collapse
|
33
|
Jiang K, Chen X, Zhao G, Wu H, Mi J, Qiu C, Peng X, Deng G. IFN-τ Plays an Anti-Inflammatory Role in Staphylococcus aureus-Induced Endometritis in Mice Through the Suppression of NF-κB Pathway and MMP9 Expression. J Interferon Cytokine Res 2017; 37:81-89. [DOI: 10.1089/jir.2016.0058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xiuying Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Junxian Mi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
34
|
Jiang KF, Zhao G, Deng GZ, Wu HC, Yin NN, Chen XY, Qiu CW, Peng XL. Polydatin ameliorates Staphylococcus aureus-induced mastitis in mice via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB pathway. Acta Pharmacol Sin 2017; 38:211-222. [PMID: 27890916 DOI: 10.1038/aps.2016.123] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/18/2016] [Indexed: 01/01/2023] Open
Abstract
Recent studies show that Polydatin (PD) extracted from the roots of Polygonum cuspidatum Sieb, a widely used traditional Chinese remedies, possesses anti-inflammatory activity in several experimental models. In this study, we investigated the anti-inflammatory effects of PD on Staphylococcus aureus-induced mastitis in mice and elucidated the potential mechanisms. In mice with S aureus-induced mastitis, administration of PD (15, 30, 45 mg/kg, ip) or dexamethasone (Dex, 5 mg/kg, ip) significantly suppressed the infiltration of inflammatory cells, ameliorated the mammary structural damage, and inhibited the activity of myeloperoxidase, a biomarker of neutrophils accumulation. Furthermore, PD treatment dose-dependently decreased the levels of TNF-α, IL-1β, IL-6 and IL-8 in the mammary gland tissues. PD treatment also dose-dependently decreased the expression of TLR2, MyD88, IRAK1, IRAK4 and TRAF6 as well as the phosphorylation of TAK1, MKK3/6, p38 MAPK, IκB-α and NF-κB in the mammary gland tissues. In mouse mammary epithelial cells (mMECs) infected by S aureus in vitro, pretreatment with PD dose-dependently suppressed the upregulated pro-inflammatory cytokines and signaling proteins, and the nuclear translocation of NF-κB p65 and AP-1. A TLR2-neutralizing antibody mimicked PD in its suppression on S aureus-induced upregulation of MyD88, p-p38 and p-p65 levels in mMECs. PD (50, 100 μg/mL) affected neither the growth of S aureus in vitro, nor the viability of mMECs. In conclusion, PD does not exhibit antibacterial activity against S aureus, its therapeutic effects in mouse S aureus-induced mastitis depend on its ability to down-regulate pro-inflammatory cytokine levels via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB signaling pathway.
Collapse
|
35
|
Catani JPP, Medrano RFV, Hunger A, Del Valle P, Adjemian S, Zanatta DB, Kroemer G, Costanzi-Strauss E, Strauss BE. Intratumoral Immunization by p19Arf and Interferon-β Gene Transfer in a Heterotopic Mouse Model of Lung Carcinoma. Transl Oncol 2016; 9:565-574. [PMID: 27916291 PMCID: PMC5143354 DOI: 10.1016/j.tranon.2016.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/29/2016] [Accepted: 09/29/2016] [Indexed: 02/06/2023] Open
Abstract
Therapeutic strategies that act by eliciting and enhancing antitumor immunity have been clinically validated as an effective treatment modality but may benefit from the induction of both cell death and immune activation as primary stimuli. Using our AdRGD-PG adenovector platform, we show here for the first time that in situ gene transfer of p19Arf and interferon-β (IFNβ) in the LLC1 mouse model of lung carcinoma acts as an immunotherapy. Although p19Arf is sufficient to induce cell death, only its pairing with IFNβ significantly induced markers of immunogenic cell death. In situ gene therapy with IFNβ, either alone or in combination with p19Arf, could retard tumor progression, but only the combined treatment was associated with a protective immune response. Specifically in the case of combined intratumoral gene transfer, we identified 167 differentially expressed genes when using microarray to evaluate tumors that were treated in vivo and confirmed the activation of CCL3, CXCL3, IL1α, IL1β, CD274, and OSM, involved in immune response and chemotaxis. Histologic evaluation revealed significant tumor infiltration by neutrophils, whereas functional depletion of granulocytes ablated the antitumor effect of our approach. The association of in situ gene therapy with cisplatin resulted in synergistic elimination of tumor progression. In all, in situ gene transfer with p19Arf and IFNβ acts as an immunotherapy involving recruitment of neutrophils, a desirable but previously untested outcome, and this approach may be allied with chemotherapy, thus providing significant antitumor activity and warranting further development for the treatment of lung carcinoma.
Collapse
Affiliation(s)
- João Paulo Portela Catani
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil
| | - Ruan F V Medrano
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil
| | - Aline Hunger
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil
| | - Paulo Del Valle
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil
| | - Sandy Adjemian
- Laboratory of Cell and Molecular Biology, Department of Immunology, Biomedical Sciences Institute, University of São Paulo, Brazil
| | - Daniela Bertolini Zanatta
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; U1138, INSERM, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Eugenia Costanzi-Strauss
- Gene Therapy Laboratory, Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, Brazil
| | - Bryan E Strauss
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of Sao Paulo/LIM 24, University of São Paulo School of Medicine, Brazil.
| |
Collapse
|
36
|
Kobayashi K, Kuki C, Oyama S, Kumura H. Pro-inflammatory cytokine TNF-α is a key inhibitory factor for lactose synthesis pathway in lactating mammary epithelial cells. Exp Cell Res 2016; 340:295-304. [DOI: 10.1016/j.yexcr.2015.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/23/2015] [Accepted: 10/24/2015] [Indexed: 10/24/2022]
|
37
|
Brazilin plays an anti-inflammatory role with regulating Toll-like receptor 2 and TLR 2 downstream pathways in Staphylococcus aureus-induced mastitis in mice. Int Immunopharmacol 2015; 27:130-7. [DOI: 10.1016/j.intimp.2015.04.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 02/06/2023]
|
38
|
Angiopoietin-like protein 2 may mediate the inflammation in murine mastitis through the activation of interleukin-6 and tumour necrosis factor-α. World J Microbiol Biotechnol 2015; 31:1235-40. [PMID: 26003652 DOI: 10.1007/s11274-015-1873-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 05/16/2015] [Indexed: 12/30/2022]
Abstract
Mastitis is the inflammation of the mammary gland. Recent research has shown that Angiopoietin-like protein 2 (ANGPTL2) is a key inflammatory mediator. In the present study, we tested whether there is a correlation between increased ANGPTL2 expression and inflammation in response to Staphylococcus aureus in murine mastitis and the mechanisms involved. Thirty mice were divided into two groups: blank control group, challenged group. The entire infused mammary glands were removed to observe the changes of histopathology, myeloperoxidase (MPO) activity, production of tumour necrosis factor-α (TNF-α) and interleukin (IL)-6, and genes expression of ANGPTL2, TNF-α and IL-6. In challenged group, the structure of mammary glands was damaged and the large areas of cell fragments were observed. The MPO activity, IL-6 and TNF-α concentrations, ANGPTL2, IL-6, and TNF-α mRNA levels were significantly elevated in challenged group compared with blank control group. The present findings indicate ANGPTL2 may mediate the inflammation in murine mastitis through the activation of IL-6 and TNF-α.
Collapse
|
39
|
Abstract
The mammary gland (MG) lacks a mucosa but is part of the mucosal immune system because of its role in passive mucosal immunity. The MG is not an inductive site for mucosal immunity. Rather, synthesis of immunoglobulin (Ig)A by plasma cells stimulated at distal inductive sites dominate in the milk of rodents, humans, and swine whereas IgG1 derived from serum predominates in ruminants. Despite the considerable biodiversity in the role of the MG, IgG passively transfers the maternal systemic immunological experience whereas IgA transfers the mucosal immunological experience. Although passive antibodies are protective, they and other lacteal constituents can be immunoregulatory. Immune protection of the MG largely depends on the innate immune system; the monocytes–macrophages group together with intraepithelial lymphocytes is dominant in the healthy gland. An increase in somatic cells (neutrophils) and various interleukins signal infection (mastitis) and a local immune response in the MG. The major role of the MG to mucosal immunity is the passive immunity supplied to the suckling neonate.
Collapse
|
40
|
Ezzat Alnakip M, Quintela-Baluja M, Böhme K, Fernández-No I, Caamaño-Antelo S, Calo-Mata P, Barros-Velázquez J. The Immunology of Mammary Gland of Dairy Ruminants between Healthy and Inflammatory Conditions. J Vet Med 2014; 2014:659801. [PMID: 26464939 PMCID: PMC4590879 DOI: 10.1155/2014/659801] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/24/2014] [Indexed: 01/01/2023] Open
Abstract
The health of dairy animals, particularly the milk-producing mammary glands, is essential to the dairy industry because of the crucial hygienic and economic aspects of ensuring production of high quality milk. Due to its high prevalence, mastitis is considered the most important threat to dairy industry, due to its impacts on animal health and milk production and thus on economic benefits. The MG is protected by several defence mechanisms that prevent microbial penetration and surveillance. However, several factors can attenuate the host immune response (IR), and the possession of various virulence and resistance factors by different mastitis-causing microorganisms greatly limits immune defences and promotes establishment of intramammary infections (IMIs). A comprehensive understanding of MG immunity in both healthy and inflammatory conditions will be an important key to understand the nature of IMIs caused by specific pathogens and greatly contributes to the development of effective control methods and appropriate detection techniques. Consequently, this review aims to provide a detailed overview of antimicrobial defences in the MG under healthy and inflammatory conditions. In this sense, we will focus on pathogen-dependent variations in IRs mounted by the host during IMI and discuss the potential ramifications of these variations.
Collapse
Affiliation(s)
- Mohamed Ezzat Alnakip
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
- Food Control Department, Dairy Division, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Al Sharkia 44519, Egypt
| | - Marcos Quintela-Baluja
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| | - Karola Böhme
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| | - Inmaculada Fernández-No
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| | - Sonia Caamaño-Antelo
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| | - Pillar Calo-Mata
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| | - Jorge Barros-Velázquez
- Department of Analytical Chemistry, Nutrition and Food Science, School of Veterinary Sciences/College of Biotechnology, University of Santiago de Compostela, Campus Lugo, Rúa Carballo Calero, 27002 Lugo, Spain
| |
Collapse
|
41
|
Xiao HB, Wang CR, Liu ZK, Wang JY. LPS induces pro-inflammatory response in mastitis mice and mammary epithelial cells: Possible involvement of NF-κB signaling and OPN. ACTA ACUST UNITED AC 2014; 63:11-6. [PMID: 25468491 DOI: 10.1016/j.patbio.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/13/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Lipopolysaccharide (LPS) has pro-inflammatory properties. This study was conducted to determine whether the LPS induced pro-inflammatory response in a model of mastitis and in mouse mammary epithelial cells (MEC). METHODS To investigate the effects of LPS in vivo, 50 μL of a solution of LPS (20 ng/μL) were infused into the mammary glands of mice. To study the effects of LPS in vitro, MEC were exposed to LPS (20 μg/mL) for 24h. Activation of nuclear factor kB (NF-κB) and myeloperoxidase (MPO) were studied. Production of pro-inflammatory cytokines (interleukin-6 [IL-6], tumor necrosis factor-alpha [TNF-alpha], interleukin-1 beta [IL-1 beta]) and expression of osteopontin (OPN) were also evaluated. RESULTS After LPS administration, route of NF-κB signaling is activated and the activity of MPO is increased. Furthermore, LPS increases the expression of OPN and production of TNF-alpha, IL-6 and IL-1 beta. CONCLUSIONS Present results demonstrate that LPS induces a pro-inflammatory response in a murine model of mastitis and suggest the involvement of the NF-κB pathway and OPN.
Collapse
Affiliation(s)
- H-B Xiao
- College of veterinary medicine, Hunan agricultural university, Changsha 410128, Furong district, China.
| | - C-R Wang
- College of veterinary medicine, Hunan agricultural university, Changsha 410128, Furong district, China
| | - Z-K Liu
- College of veterinary medicine, Hunan agricultural university, Changsha 410128, Furong district, China
| | - J-Y Wang
- Huai Hua vocational and technical college, department of animal science and technology, Huaihua 418000, China
| |
Collapse
|
42
|
Breyne K, Cool SK, Demon D, Demeyere K, Vandenberghe T, Vandenabeele P, Carlsen H, Van Den Broeck W, Sanders NN, Meyer E. Non-classical proIL-1beta activation during mammary gland infection is pathogen-dependent but caspase-1 independent. PLoS One 2014; 9:e105680. [PMID: 25162221 PMCID: PMC4146512 DOI: 10.1371/journal.pone.0105680] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/22/2014] [Indexed: 01/15/2023] Open
Abstract
Infection of the mammary gland with live bacteria elicits a pathogen-specific host inflammatory response. To study these host-pathogen interactions wild type mice, NF-kappaB reporter mice as well as caspase-1 and IL-1beta knockout mice were intramammarily challenged with Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The murine mastitis model allowed to compare the kinetics of the induced cytokine protein profiles and their underlying pathways. In vivo and ex vivo imaging showed that E. coli rapidly induced NF-kappaB inflammatory signaling concomitant with high mammary levels of TNF-alpha, IL-1 alpha and MCP-1 as determined by multiplex analysis. In contrast, an equal number of S. aureus bacteria induced a low NF-kappaB activity concomitant with high mammary levels of the classical IL-1beta fragment. These quantitative and qualitative differences in local inflammatory mediators resulted in an earlier neutrophil influx and in a more extensive alveolar damage post-infection with E. coli compared to S. aureus. Western blot analysis revealed that the inactive proIL-1beta precursor was processed into pathogen-specific IL-1beta fragmentation patterns as confirmed with IL-1beta knockout animals. Additionally, caspase-1 knockout animals allowed to investigate whether IL-1beta maturation depended on the conventional inflammasome pathway. The lack of caspase-1 did not prevent extensive proIL-1beta fragmentation by either of S. aureus or E. coli. These non-classical IL-1beta patterns were likely caused by different proteases and suggest a sentinel function of IL-1beta during mammary gland infection. Thus, a key signaling nodule can be defined in the differential host innate immune defense upon E. coli versus S. aureus mammary gland infection, which is independent of caspase-1.
Collapse
Affiliation(s)
- Koen Breyne
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- * E-mail:
| | - Steven K. Cool
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Dieter Demon
- Department of Medical Protein Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent University, Ghent, Belgium
| | - Kristel Demeyere
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Tom Vandenberghe
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent University, Zwijnaarde, Belgium
| | - Peter Vandenabeele
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent University, Zwijnaarde, Belgium
| | - Harald Carlsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Wim Van Den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N. Sanders
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evelyne Meyer
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
43
|
Wang Y, He X, Hao D, Yu D, Liang J, Qu Y, Sun D, Yang B, Yang K, Wu R, Wang J. Low-level laser therapy attenuates LPS-induced rats mastitis by inhibiting polymorphonuclear neutrophil adhesion. J Vet Med Sci 2014; 76:1443-50. [PMID: 25452258 PMCID: PMC4272976 DOI: 10.1292/jvms.14-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to
investigate the effects of low-level laser therapy (LLLT) on a rat model of
lipopolysaccharide (LPS)-induced mastitis and its underlying molecular mechanisms. The rat
model of mastitis was induced by inoculation of LPS through the canals of the mammary
gland. The results showed that LPS-induced secretion of IL-1β and IL-8 significantly
decreased after LLLT (650 nm, 2.5 mW, 30 mW/cm2). LLLT also
inhibited intercellular adhesion molecule-1 (ICAM-1) expression and attenuated the
LPS-induced decrease of the expression of CD62L and increase of the expression of CD11b.
Moreover, LLLT also suppressed LPS-induced polymorphonuclear neutrophils (PMNs) entering
the alveoli of the mammary gland. The number of PMNs in the mammary alveolus and the
myeloperoxidase (MPO) activity were decreased after LLLT. These results suggested that
LLLT therapy is beneficial in decreasing the somatic cell count and improving milk
nutritional quality in cows with an intramammary infection.
Collapse
Affiliation(s)
- Yueqiang Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ingman WV, Glynn DJ, Hutchinson MR. Inflammatory mediators in mastitis and lactation insufficiency. J Mammary Gland Biol Neoplasia 2014; 19:161-7. [PMID: 24961655 DOI: 10.1007/s10911-014-9325-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/18/2014] [Indexed: 12/15/2022] Open
Abstract
Mastitis is a common inflammatory disease during lactation that causes reduced milk supply. A growing body of evidence challenges the central role of pathogenic bacteria in mastitis, with disease severity associated with markers of inflammation rather than infection. Inflammation in the mammary gland may be triggered by microbe-associated molecular patterns (MAMPs) as well as danger-associated molecular patterns (DAMPs) binding to pattern recognition receptors such as the toll-like receptors (TLRs) on the surface of mammary epithelial cells and local immune cell populations. Activation of the TLR4 signalling pathway and downstream nuclear factor kappa B (NFkB) is critical to mediating local mammary gland inflammation and systemic immune responses in mouse models of mastitis. However, activation of NFkB also induces epithelial cell apoptosis and reduced milk protein synthesis, suggesting that inflammatory mediators activated during mastitis promote partial involution. Perturbed milk flow, maternal stress and genetic predisposition are significant risk factors for mastitis, and could lead to a heightened TLR4-mediated inflammatory response, resulting in increased susceptibility and severity of mastitis disease in the context of low MAMP abundance. Therefore, heightened host inflammatory signalling may act in concert with pathogenic or commensal bacterial species to cause both the inflammation associated with mastitis and lactation insufficiency. Here, we present an alternate paradigm to the widely held notion that breast inflammation is driven principally by infectious bacterial pathogens, and suggest there may be other therapeutic strategies, apart from the currently utilised antimicrobial agents, that could be employed to prevent and treat mastitis in women.
Collapse
Affiliation(s)
- Wendy V Ingman
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Woodville, Australia
| | | | | |
Collapse
|
45
|
Glynn DJ, Hutchinson MR, Ingman WV. Toll-like receptor 4 regulates lipopolysaccharide-induced inflammation and lactation insufficiency in a mouse model of mastitis. Biol Reprod 2014; 90:91. [PMID: 24671877 DOI: 10.1095/biolreprod.114.117663] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lactation mastitis is a debilitating inflammatory breast disease in postpartum women. Disease severity is associated with markers of inflammation rather than bacterial load, suggesting that immune-signaling pathways activated in the host are important in the disease pathology. The role of the innate pattern recognition receptor toll-like receptor 4 (TLR4) in progression and resolution of mastitislike disease was investigated in a mouse model. Lipopolysaccharide in Matrigel (10 μg/10 μl) was administered into the teat canal of lactating Tlr4 null mutant and wild-type mice to induce a localized area of inflammation. Mastitis induction resulted in a marked influx of RB6-positive neutrophils and F4/80-positive macrophages, which was higher in Tlr4(-/-) mice compared to wild-type mice. Tlr4 null mutation resulted in an altered immune-signaling fingerprint following induction of mastitis, with attenuated serum cytokines, including CXCL1, CCL2, interleukin 1 beta, and tumor necrosis factor alpha compared to wild-type mice. In both genotypes, the localized area of inflammation had resolved after 7 days, and milk protein was evident. However, the mammary glands of wild-type mice exhibited reduced capacity for milk production, with decreased percent area populated with glandular epithelium and decreased abundance of nuclear phosphorylated signal transducer and activator of transcription 5 compared to Tlr4 null mice. This study demonstrates that inflammatory pathways activated in the host are critically important in mastitis disease progression and suggests that lactation insufficiency associated with mastitis may be a consequence of TLR4-mediated inflammation, rather than the bacterial infection itself.
Collapse
Affiliation(s)
- Danielle J Glynn
- School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia, Australia
| | | | | |
Collapse
|
46
|
Baicalin inhibits Staphylococcus aureus-induced apoptosis by regulating TLR2 and TLR2-related apoptotic factors in the mouse mammary glands. Eur J Pharmacol 2014; 723:481-8. [DOI: 10.1016/j.ejphar.2013.10.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/10/2013] [Accepted: 10/17/2013] [Indexed: 01/04/2023]
|
47
|
Seroussi E, Klompus S, Silanikove M, Krifucks O, Shapiro F, Gertler A, Leitner G. Nonbactericidal secreted phospholipase A2s are potential anti-inflammatory factors in the mammary gland. Immunogenetics 2013; 65:861-71. [PMID: 24091988 DOI: 10.1007/s00251-013-0738-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 09/20/2013] [Indexed: 12/17/2022]
Abstract
The recent burst of duplication and divergence of the bovine PLA2G2D genes is considered typical of immune response genes, and it was recently shown that PLA2G2D is abundantly expressed in mouse leukocytes and acts as an immunosuppressive phospholipase. Analysis of 1,143 Holstein bulls indicated that the four common haplotypes spanning PLA2G2D display copy number variation ranging from 1 to 4 per haploid genome. Association of the fourth haplotype with negative total merit remained significant (P < 0.002) when corrected for population relatedness. We compared the lipase and bactericidal activities of bovine pancreatic PLA2G1B with human PLA2G2A and G2D and bovine PLA2G2D1 and G2D4 proteins, which had been subcloned, expressed, and refolded by us, and the impact of point mutations in the calcium binding site was investigated. All tested phospholipases were ineffective bactericides of Escherichia coli isolated from bovine mastitis. However, in lactating mice treated with E. coli or lipopolysaccharide (LPS), intramammary injection of bovine PLA2G1B relieved visual and histological inflammation and reduced blood levels of infiltrating lactose. Further studies are warranted to determine whether the observed anti-inflammatory effect involves competitive binding of the receptor Pla2r1 which may mimic the LPS resistance effect in Pla2r1-deficient mice.
Collapse
Affiliation(s)
- Eyal Seroussi
- Institute of Animal Science, ARO, The Volcani Center, P.O. Box 6, Beit Dagan, 50250, Israel,
| | | | | | | | | | | | | |
Collapse
|
48
|
Li F, Liang D, Yang Z, Wang T, Wang W, Song X, Guo M, Zhou E, Li D, Cao Y, Zhang N. Astragalin suppresses inflammatory responses via down-regulation of NF-κB signaling pathway in lipopolysaccharide-induced mastitis in a murine model. Int Immunopharmacol 2013; 17:478-82. [PMID: 23928506 DOI: 10.1016/j.intimp.2013.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 07/07/2013] [Accepted: 07/21/2013] [Indexed: 12/19/2022]
Abstract
Mastitis is a prevalent and economic disease around the world and defined as infection and inflammation of the mammary gland. Astragalin, a bioactive component isolated from persimmon or Rosa agrestis, has been reported to have anti-inflammatory properties. To investigate the potential therapeutic effect of astragalin in mastitis, a murine model of mastitis was induced by administration of LPS in mammary gland. Astragalin was applied 1h before and 12h after LPS treatment. The results showed that astragalin attenuated the infiltration of inflammatory cells, the activity of myeloperoxidase (MPO) and the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) in a dose-dependent manner. Additionally, Western blotting results showed that astragalin efficiently blunt decreased nuclear factor-kappaB (NF-κB) activation by inhibiting the degradation and phosphorylation of IκBα and the nuclear translocation of p65. These results suggested that astragalin exerts anti-inflammatory properties in LPS-mediated mastitis, possibly through inhibiting inhibition of the NF-κB signaling pathway, which mediates the expression of pro-inflammatory cytokines. Astragalin may be a potential therapeutic agent against mastitis.
Collapse
Affiliation(s)
- Fengyang Li
- Department of Clinical Veterinary Medicine, Jilin University, Changchun, Jilin Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Histological analysis of mammary gland remodeling caused by lipopolysaccharide in lactating mice. Cell Tissue Res 2013; 354:495-506. [DOI: 10.1007/s00441-013-1688-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/25/2013] [Indexed: 12/20/2022]
|
50
|
Mintz M, Mintz D, Ilia-Ezra R, Shpigel NY. Pam3CSK4/TLR2 signaling elicits neutrophil recruitment and restricts invasion of Escherichia coli P4 into mammary gland epithelial cells in a murine mastitis model. Vet Immunol Immunopathol 2013; 152:168-75. [DOI: 10.1016/j.vetimm.2012.09.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|