1
|
Im J, Jeon JH, Lee D, Park JW, Jun W, Lim S, Park OJ, Yun CH, Han SH. Muramyl dipeptide potentiates Staphylococcus aureus lipoteichoic acid-induced nitric oxide production via TLR2/NOD2/PAFR signaling pathways. Front Immunol 2024; 15:1451315. [PMID: 39712020 PMCID: PMC11659290 DOI: 10.3389/fimmu.2024.1451315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
Lipoteichoic acid (LTA) and peptidoglycan (PGN) are considered as key virulence factors of Staphylococcus aureus, which is a representative sepsis-causing Gram-positive pathogen. However, cooperative effect of S. aureus LTA and PGN on nitric oxide (NO) production is still unclear despite the pivotal roles of NO in initiation and progression of sepsis. We here evaluated the cooperative effects of S. aureus LTA (SaLTA) and muramyl dipeptide (MDP), the minimal structure of PGN, on NO production in both a mouse macrophage-like cell line, RAW 264.7 and mouse bone marrow-derived macrophages (BMMs). Although MDP alone did not affect NO production, MDP potently enhanced SaLTA-induced NO production via the expression of inducible NO synthases. The enhanced NO production was ameliorated in BMMs from TLR2-, CD14-, MyD88-, and NOD2-deficient mice. Moreover, the augmented SaLTA-induced NO production by MDP was attenuated by inhibitors specific for PAFR and MAP kinases. Furthermore, MDP also potently increased SaLTA-induced activities of STAT1, NF-κB, and AP-1 transcription factors, and specific inhibitors for these transcription factors suppressed the elevated NO production. Collectively, these results demonstrated that MDP potentiates SaLTA-induced NO production via TLR2/NOD2/PAFR, MAP kinases signaling axis, resulting in the activation of NF-κB, AP-1 and STAT1 transcription factors.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jun Ho Jeon
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jeong Woo Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Woohyung Jun
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Suwon Lim
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Institutes of Green-bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Elahirad E, Gharagozlou MJ, Khosravi A, Sasani F. TLRs expression in canine mammary gland neoplasms: a pathological and molecular study. Vet Immunol Immunopathol 2023; 261:110611. [PMID: 37245345 DOI: 10.1016/j.vetimm.2023.110611] [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: 03/07/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/30/2023]
Abstract
TLRs are a class of PRRs that play a vital role in innate immunity. TLRs are expressed on immune cells and mammary epithelial cells. They can promote tumor growth, angiogenesis, invasion, and viability signaling. The current study aimed to test the correlation between histologic types and grades of neoplasms and TLRs gene expression levels. Twenty-one tissue samples of canine mammary neoplasms were stained with H&E. Then, it evaluated histologic type and grade according to the methods of Goldschmidt et al. and Peña, respectively. We established real-time PCR quantification assays to measure the mRNA abundances of TLRs in normal and neoplastic mammary glands. Profile pattern of TLR 1, 2, 3, 4, 5, 6, and 9 genes expression in canine mammary glands performed in 21 samples of mammary gland neoplasms and three non-neoplastic mammary gland samples from normal dogs. TLR 3, 4, and 9 mRNA overexpression were detected. In addition, tubulopapillary carcinoma grade II, SCC grade III, and carcinoma mixed type grade II demonstrated the highest relative TLR-3, and 9 mRNA expression levels. Complex carcinoma grade I, ductal carcinoma grade II, and anaplastic carcinoma grade II showed the highest relative TLR4 mRNA expression level. Although histopathological characteristics of tumors, including histologic type, grade, and inflammation, influenced TLRs mRNA expression level, such correlation was insignificant (P > 0.05).
Collapse
Affiliation(s)
- Elnaz Elahirad
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | | | - Alireza Khosravi
- Mycology Research Center, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farhang Sasani
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
3
|
Xu P, Xu X, Fotina H, Fotina T. Anti-inflammatory effects of chlorogenic acid from Taraxacum officinale on LTA-stimulated bovine mammary epithelial cells via the TLR2/NF-κB pathway. PLoS One 2023; 18:e0282343. [PMID: 36947494 PMCID: PMC10032541 DOI: 10.1371/journal.pone.0282343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/13/2023] [Indexed: 03/23/2023] Open
Abstract
Mastitis is an inflammatory disease caused by microbial infection. Chlorogenic acid (CGA), one of the major phenolic acids in Taraxacum officinale, has natural antioxidant and anti-inflammatory properties in various cell types; however, the effects of CGA on Lipoteichoic acid (LTA)-induced bovine mammary epithelial cells (BMECs) have not been investigated. In this study, the CGA content in T. officinale was determined by High-performance liquid chromatography (HPLC). BMECs were infected with LTA to induce the mastitis model. Different concentrations of CGA were administered after establishing the LTA infection. The results showed that the T. officinale contained CGA 1.36 mg/g. CGA significantly reduced the pro-inflammatory gene and protein expression of TNF-α, IL-6, and IL-1β. In addition, CGA downregulated the NO, TLR2, and NF-κB signaling pathways in LTA-infected bovine mammary epithelial cells. Our results indicate that CGA reduced the expression of TNF-α, IL-6, IL-1β, and TLR2 by inhibiting the phosphorylation of proteins in the NF-κB signaling pathways in a dose-dependent manner. This finding suggests that CGA may be a potential agent for the treatment of mastitis in dairy cows.
Collapse
Affiliation(s)
- Ping Xu
- School of Life Science and Basic Medicine, Xinxiang University, Xinxiang, China
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Xiaobo Xu
- School of Life Science and Basic Medicine, Xinxiang University, Xinxiang, China
| | - Hanna Fotina
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Tetiana Fotina
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| |
Collapse
|
4
|
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: 19] [Impact Index Per Article: 6.3] [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
|
5
|
Dong W, Chen Y, Zhang Q, Zhao X, Liu P, He H, Lu T, He Y, Du X, Hu J, Zhao X, Zhang Y. Effects of lipoteichoic and arachidonic acids on the immune-regulatory mechanism of bovine mammary epithelial cells using multi-omics analysis. Front Vet Sci 2022; 9:984607. [PMID: 36090174 PMCID: PMC9450935 DOI: 10.3389/fvets.2022.984607] [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: 07/02/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus is one of the most important pathogens causing mastitis in dairy cows. It mainly utilizes the properties of its pathogenic factor, lipoteichoic acid (LTA), to elicit a host-cell inflammatory response and evade the host-cell immune response. Arachidonic acid (AA) has a regulatory role in the inflammatory response, cell metabolism, and apoptosis. The study aimed to establish a cell model by determining the optimal concentration of LTA and AA for cell induction using the Cell Counting Kit−8 assay and the quantitative polymerase chain reaction of interleukin (IL)-1β, IL-2, and IL-6. MAC-T cells were planted in 36 10-cm2 culture dishes at a density of 1 × 107 cells per dish. They were treated with LTA for 24 h to constitute the LTA group and with AA for 12 h to constitute the AA group. The cells were pretreated with LTA for 24 h followed by treatment with AA for 12 h to constitute the LTA + AA group. Using proteomic, transcriptomic, and metabolomic analyses, this study determined that LTA can regulate the expression of Actin Related protein 2/3 complex (ARPC)3, ARPC4, Charged Multivesicular Body Protein 3, protein kinase cGMP-dependent, NF-κB Inhibitor Alpha,and other genes to affect cellular metabolism, immune regulation and promote apoptosis. In contrast, AA was observed to regulate the expression of genes such as ARPC3, ARPC4, Charged Multivesicular Body Protein 3, Laminin Gamma 1, Insulin Receptor, Filamin B, and Casein Kinase 1 Epsilon to inhibit cellular apoptosis and promote immune regulation, which provides a theoretical basis for future studies.
Collapse
Affiliation(s)
- Weitao Dong
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Yan Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Quanwei Zhang
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoxuan Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Peiwen Liu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Haijian He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Ting Lu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Yuxuan He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Xianghong Du
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation of Gansu Province, Lanzhou, China
- *Correspondence: Yong Zhang
| |
Collapse
|
6
|
Hommes JW, Surewaard BGJ. Intracellular Habitation of Staphylococcus aureus: Molecular Mechanisms and Prospects for Antimicrobial Therapy. Biomedicines 2022; 10:1804. [PMID: 36009351 PMCID: PMC9405036 DOI: 10.3390/biomedicines10081804] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/23/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections pose a global health threat, especially with the continuous development of antibiotic resistance. As an opportunistic pathogen, MRSA infections have a high mortality rate worldwide. Although classically described as an extracellular pathogen, many studies have shown over the past decades that MRSA also has an intracellular aspect to its infectious cycle, which has been observed in vitro in both non-professional as well as professional phagocytes. In vivo, MRSA has been shown to establish an intracellular niche in liver Kupffer cells upon bloodstream infection. The staphylococci have evolved various evasion strategies to survive the antimicrobial environment of phagolysosomes and use these compartments to hide from immune cells and antibiotics. Ultimately, the host cells get overwhelmed by replicating bacteria, leading to cell lysis and bacterial dissemination. In this review, we describe the different intracellular aspects of MRSA infection and briefly mention S. aureus evasion strategies. We discuss how this intracellular niche of bacteria may assist in antibiotic tolerance development, and lastly, we describe various new antibacterial strategies that target the intracellular bacterial niche.
Collapse
Affiliation(s)
| | - Bas G. J. Surewaard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| |
Collapse
|
7
|
Arbab AAI, Lu X, Abdalla IM, Idris AA, Chen Z, Li M, Mao Y, Xu T, Yang Z. Metformin Inhibits Lipoteichoic Acid-Induced Oxidative Stress and Inflammation Through AMPK/NRF2/NF-κB Signaling Pathway in Bovine Mammary Epithelial Cells. Front Vet Sci 2021; 8:661380. [PMID: 34262962 PMCID: PMC8274905 DOI: 10.3389/fvets.2021.661380] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
The objective of this research was to explore the effect of metformin on the lipoteichoic acid (LTA)–induced mastitis model using isolated primary bovine mammary epithelial cells (PBMECs). The PBMECs were exposed to either 3 mM metformin for 12 h as a metformin group (MET) or 100 μg/mL LTA for 6 h as LTA group (LTA). Cells pretreated with 3 mM metformin for 12 h followed by washing and 100 μg/mL LTA exposure for 6 h served as the MET + LTA group. Phosphate-buffered saline was added to cells as the control group. PBMECs pretreated with different metformin doses were analyzed by a flow cytometry (annexin V–fluorescein isothiocyanate assay) to detect the cell apoptotic rate. We performed quantitative reverse transcriptase–polymerase chain reaction and Western blot analysis to evaluate the inflammatory and oxidative responses to metformin and LTA by measuring cellular cytotoxicity, mRNA expression, and protein expression. Immunofluorescence was used to evaluate nuclear localization. The results showed that the gene expression of COX2, IL-1β, and IL-6 significantly increased in the cells challenged with LTA doses compared to control cells. In inflammatory PBMECs, metformin attenuated LTA-induced expression of inflammatory genes nuclear factor κB (NF-κB) p65, tumor necrosis factor α, cyclooxygenase 2, and interleukin 1β, as well as the nuclear localization and phosphorylation of NF-κBp65 protein, but increased the transcription of nuclear factor erythroid 2–related factor 2 (Nrf2) and Nrf2-targeted antioxidative genes heme oxygenase-1 (HO-1) and Gpx1, as well as the nuclear localization of HO-1 protein. Importantly, metformin-induced activation of Nrf2 is AMP-activated protein kinase (AMPK)–dependent; as metformin-pretreated PBMECs activated AMPK signaling via the upregulation of phosphorylated AMPK levels, cell pretreatment with metformin also reversed the translocation of Nrf2 that was LTA inhibited. This convergence between AMPK and Nrf2 pathways is essential for the anti-inflammatory effect of metformin in LTA-stimulated PBMECs. Altogether, our results indicate that metformin exerts anti-inflammation and oxidative stress through regulation of AMPK/Nrf2/NF-κB signaling pathway, which highlights the role of AMPK as a potential therapeutic strategy for treatment of bovine mastitis.
Collapse
Affiliation(s)
- Abdelaziz Adam Idriss Arbab
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Darfur College, Biomedical Research Institute, Niyla, Sudan
| | - Xubin Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | | | | | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Mingxun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yongjiang Mao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tianle Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Zhangping Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
8
|
Cytotoxicity and anti-inflammatory effect of a novel diminazene aceturate derivative in bovine mammary epithelial cells. Res Vet Sci 2021; 137:102-110. [PMID: 33964615 DOI: 10.1016/j.rvsc.2021.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
Diminazene aceturate (DA) has been used in the treatment of infections of trypanosomes in animals. Interestingly, its anti-inflammatory effect has recently gained increased interests. However, DA has been reported to have toxic side effects that limit its application. Therefore, we synthesized and screened a novel low-toxic DA derivative, namely the DA derivative 3 (DAD3). In the present study, anti-inflammatory effect of DAD3 was evaluated bovine mammary epithelial cells (BMECs) in vitro model. The results demonstrated that DAD3 had less cytotoxicity, and had a stronger effect in inhibiting secretion of inflammatory factors in BMECs, compared to DA. Mechanistically, DAD3 was able to inhibit the production of pro-inflammatory factors in part by suppressing the generation of mitochondrial reactive oxygen species (ROS) in BMECs upon LPS stimulation. Molecular analysis further indicated that DAD3 was capable of resolving inflammation in BMECs through a mechanism by preventing nuclear translocation of NF-p65, subsequently inhibiting transcription of inflammatory factors. In this context, DAD3 inhibited the phosphorylation of IκB, ERK, JNK and P-38 proteins of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. These results suggested the DAD3 was a novel DA derivative with low toxicity and strong anti-inflammatory effects in BMECs exposed to LPS, through a mechanism by blocking the NF-κB and MAPK signaling pathways. This study also provides an evidence that the DAD3 may be a novel anti-inflammatory agents warranted for further investigation in treatment of mastitis in cows.
Collapse
|
9
|
Kirsanova E, Heringstad B, Lewandowska-Sabat A, Olsaker I. Identification of candidate genes affecting chronic subclinical mastitis in Norwegian Red cattle: combining genome-wide association study, topologically associated domains and pathway enrichment analysis. Anim Genet 2019; 51:22-31. [PMID: 31808564 DOI: 10.1111/age.12886] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Abstract
The aim of this study was to identify genes associated with chronic subclinical mastitis (SCM) in Norwegian Red (NR) cattle. Twelve SCM traits defined based on fixed threshold for test-day somatic cell count (SCC) were, together with lactation-average somatic cell score (LSCS) used for association and pathway enrichment analyses. A GWAS was performed on 3795 genotyped NR bulls with 777K SNP data and phenotypic information from 7 300 847 test-day SCC observations from 3 543 764 cows. At 5% chromosome-wide significance level 36 unique SNP were detected to be associated with one or more of the traits. These SNPs were analysed for linked genes using genomic positions of topologically associated domains (TAD). For the SCM traits with SCC >50 000 and >100 000 cells/ml on two test-days in a row and LSCS, the same top significant genes were identified - checkpoint clamp loader component (RAD17) and cyclin B1 (CCNB1). The SCM traits with SCC >250 000, 300 000, 350 000 or 400 000 cells/ml on two test-days in a row and D400 (number of days before the first case with SCC >400 000 cells/ml) displayed similar top significant genes: acyl-CoA thioesterase 2 and 4 (ACOT2; ACOT4). For the traits SCM200_3 (SCC >200 000 cells/ml on three test-days in a row) and SCM150, SCM200 (SCC >150 000; 200 000 cells/ml on two test-days in a row) a group of chemokine (C-X-C motif) ligand genes and the Fos proto-oncogene, AP-1 transcription factor subunit (FOS) gene, were identified. Further functional studies of these identified candidate genes are necessary to clarify their actual role in development of chronic SCM in NR cattle.
Collapse
Affiliation(s)
- E Kirsanova
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - B Heringstad
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Oslo, Norway.,Geno Breeding and A.I. Association, Hamar, Norway
| | - A Lewandowska-Sabat
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - I Olsaker
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
10
|
Gomez DE, Galvão KN, Rodriguez-Lecompte JC, Costa MC. The Cattle Microbiota and the Immune System: An Evolving Field. Vet Clin North Am Food Anim Pract 2019; 35:485-505. [PMID: 31590899 DOI: 10.1016/j.cvfa.2019.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
New insights into the host-microbiota relationship have recently emerged with the advancement of molecular technologies such as next-generation sequencing. This article presents the current knowledge regarding the interaction between bacteria and the immune system of the gut, the uterus, and the mammary gland of cattle.
Collapse
Affiliation(s)
- Diego E Gomez
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 Southwest 16th Avenue, Gainesville, FL 32608, USA.
| | - Klibs N Galvão
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 Southwest 16th Avenue, Gainesville, FL 32608, USA
| | - Juan C Rodriguez-Lecompte
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3, Canada
| | - Marcio C Costa
- Department of Veterinary Biomedicine, University of Montreal, 3200 Rue Sicotte, Saint-Hyacinthe, Quebec J2S 2M2, Canada
| |
Collapse
|
11
|
Wang X, Zhang M, Jiang N, Zhang A. Sodium Phenylbutyrate Ameliorates Inflammatory Response Induced by Staphylococcus aureus Lipoteichoic Acid via Suppressing TLR2/NF-κB/NLRP3 Pathways in MAC-T Cells. Molecules 2018; 23:molecules23123056. [PMID: 30469547 PMCID: PMC6321250 DOI: 10.3390/molecules23123056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023] Open
Abstract
This study aimed to investigate the anti-inflammatory properties of sodium phenylbutyrate (SPB) against Staphylococcus aureus (S. aureus) lipoteichoic acid (LTA)-stimulated bovine mammary alveolar (MAC-T) cells. Quantitative PCR was performed to examine the effect of SPB on inflammatory cytokines and host defense peptide (HDP) gene expression. Western blot wanalysis was used to detect the effect of SPB on the TLR2/NF-κB/NLRP3 signaling pathway. The results showed that SPB significantly suppressed the expression of TNF-α, IL-1β, IL-6; meanwhile, the markedly decreased expression of LTA-stimulated TLR2, NLRP3, ASC, caspase-1, and IL-1β, and the inhibited IkBα and p65 phosphorylation were also observed. However, increased TAP and Bac5 expression in LTA-stimulated MAC-T cells was further detected. In summary, these results suggest that SPB ameliorates the inflammatory response induced by S. aureus LTA via suppressing the TLR2/NF-κB/NLRP3 signaling pathway, which indicates that SPB may be a potential agent for the treatment of bovine mastitis.
Collapse
Affiliation(s)
- Xin Wang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Mengmeng Zhang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Ning Jiang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| | - Aizhong Zhang
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Heilongjiang, Daqing 163319, China.
| |
Collapse
|
12
|
Rainard P, Foucras G. A Critical Appraisal of Probiotics for Mastitis Control. Front Vet Sci 2018; 5:251. [PMID: 30364110 PMCID: PMC6191464 DOI: 10.3389/fvets.2018.00251] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/19/2018] [Indexed: 01/13/2023] Open
Abstract
The urge to reduce antimicrobials use in dairy farming has prompted a search for alternative solutions. As infections of the mammary gland is a major reason for antibiotic administration to dairy ruminants, mammary probiotics have recently been presented as a possible alternative for the treatment of mastitis. To assess the validity of this proposal, we performed a general appraisal of the knowledge related to probiotics for mammary health by examining their potential modes of action and assessing the compatibility of these mechanisms with the immunobiology of mammary gland infections. Then we analyzed the literature published on the subject, taking into account the preliminary in vitro experiments and the in vivo trials. Preliminary experiments aimed essentially at exploring in vitro the capacity of putative probiotics, mainly lactic acid bacteria (LABs), to interfere with mastitis-associated bacteria or to interact with mammary epithelial cells. A few studies used LABs selected on the basis of bacteriocin production or the capacity to adhere to epithelial cells to perform in vivo experiments. Intramammary infusion of LABs showed that LABs are pro-inflammatory for the mammary gland, inducing an intense influx of neutrophils into milk during lactation and at drying-off. Yet, their capacity to cure mastitis remains to be established. A few preliminary studies tackle the possibility of using probiotics to interfere with the teat apex microbiota or to prevent the colonization of the teat canal by pathogenic bacteria. From the analysis of the published literature, it appears that currently there is no sound scientific foundation for the use of probiotics to prevent or treat mastitis. We conclude that the prospects for oral probiotics are not promising for ruminants, those for intramammary probiotics should be considered with caution, but that teat apex probiotics deserve further research.
Collapse
Affiliation(s)
- Pascal Rainard
- ISP, INRA, Université de Tours, UMR 1282, Nouzilly, France
| | - Gilles Foucras
- IHAP, Université de Toulouse, ENVT, INRA, UMR1225, Toulouse, France
| |
Collapse
|
13
|
Silva LG, Ferguson BS, Faciola AP. Rapid Communication: Prolactin and hydrocortisone impact TNFα-mediated mitogen-activated protein kinase signaling and inflammation of bovine mammary epithelial (MAC-T) cells. J Anim Sci 2018; 95:5524-5531. [PMID: 29293766 DOI: 10.2527/jas2017.2028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The objective of this study was to evaluate the effects of the hormones prolactin (PRL) and hydrocortisone (HC) on bovine mammary alveolar (MAC-T) cells mitogen-activated protein kinase (MAPK) inflammatory signaling and inflammatory gene expression. MAC-T cells were cultured in the presence (+PRL +HC; Dulbecco's modified Eagle's medium [DMEM] 10% fetal bovine serum, 10 µg/mL of insulin, 100 IU/mL penicillin, 100 µg/mL streptomycin, 1 µg/mL ovine PRL, 0.5 µg/mL HC, and 10 m sodium acetate) or the absence (-PRL -HC; DMEM 10% fetal bovine serum, 10 µg/mL insulin, 100 IU/mL penicillin , and 100 µg/mL streptomycin) of PRL and HC, and MAPK (extracellular signal-regulated kinase [ERK], c-Jun N-terminal kinase [JNK], and p38) phosphorylation and inflammatory gene expression were examined in response to tumor necrosis factor α (TNFα). Statistical analysis was assessed using 1-way ANOVA, and Tukey's post hoc analysis was used to assess statistical significance when ≤ 0.05. MAC-T cells cultured in +PRL +HC and -PRL -HC were co-stimulated with increasing concentrations of TNFα (0, 10, 30, 100, 300, and 1,000 p). Cell lysates were harvested 15 min after TNFα stimulation and assessed for MAPK phosphorylation using immunoblotting. c-Jun N-terminal kinase and p38 phosphorylation increased in a dose-dependent manner and was greater in cells cultured in -PRL -HC. MAC-T cells cultured in +PRL +HC and -PRL -HC were next stimulated with TNFα (300 p), and lysates were harvested over time (0, 15, 30, 60, 120, and 180 min) after TNFα stimulation. c-Jun N-terminal kinase and p38 phosphorylation was transiently increased in MAC-T cells stimulated with TNFα; however, JNK and p38 signaling was greater in MAC-T cells cultured in -PRL -HC. We next examined inflammatory gene expression in MAC-T cells cultured in +PRL +HC and -PRL -HC. Cells were co-stimulated with (300 p) or without TNFα. Ribonucleic acid was isolated 1 h after TNFα stimulation, and a PCR array was performed to examine the expression of 83 inflammatory genes. Gene expression was increased in MAC-T cells in response to TNFα. Consistent with enhanced MAPK signaling, inflammatory gene expression was increased in MAC-T cells cultured in -PRL -HC. Real-time quantitative PCR of 6 target genes was used to validate the PCR array findings. Collectively, our data demonstrate that -PRL -HC MAC-T cells are more responsive to TNFα stimuli. These findings suggest that cell culture conditions (e.g., treatment with hormones) greatly impact cellular response and should be considered prior to experimental design and hypothesis testing.
Collapse
|
14
|
Rainard P, Foucras G, Boichard D, Rupp R. Invited review: Low milk somatic cell count and susceptibility to mastitis. J Dairy Sci 2018; 101:6703-6714. [PMID: 29803421 DOI: 10.3168/jds.2018-14593] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/13/2018] [Indexed: 12/17/2022]
Abstract
An enduring controversy exists about low milk cell counts and susceptibility to mastitis. The concentration of milk leukocytes, or somatic cell count (SCC), is a well-established direct indicator of mammary gland inflammation that is highly correlated with the presence of a mammary infection. The SCC is also used as a trait for the selection of dairy ruminants less prone to mastitis. As selection programs favor animals with less SCC, and as milk cells contribute to the defense of the mammary gland, the idea that susceptibility to mastitis could possibly be increased in the long term has been put forward and is still widely debated. Epidemiological and experimental studies aimed at relating SCC to susceptibility to mastitis have yielded results that seem contradictory at first sight. Nevertheless, by taking into account the immunobiology of milk and mammary tissue cells and their role in the defense against infection, along with recent studies on SCC-based divergent selection of animals, the issue can be settled. Apparent SCC-linked susceptibility to mastitis is a phenotypic trait that may be linked to immunomodulation but not to selection.
Collapse
Affiliation(s)
- P Rainard
- ISP, Université de Tours, INRA, UMR1282, F-37380 Nouzilly, France.
| | - G Foucras
- IHAP, Université de Toulouse, ENVT, INRA, UMR1225, F-31076 Toulouse, France
| | - D Boichard
- GABI, INRA, AgroParisTech, Université Paris Saclay, F-78350 Jouy-en-Josas, France
| | - R Rupp
- GenPhySE, Université de Toulouse, INRA, F-31320 Castanet-Tolosan, France
| |
Collapse
|
15
|
Sulabh S, Panigrahi M, Ahmad SF, Varshney R, Verma A, Baba NA, Kumar S, Kumari S, Chauhan A, Kumar P, Bhushan B. Peptidoglycan and Lipoteichoic Acid Induces Differential mRNA Response of Immune-Related Genes in PBMC of Crossbred, Tharparkar Cattle and Murrah Buffalo. Anim Biotechnol 2018; 30:166-174. [DOI: 10.1080/10495398.2018.1461633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sourabh Sulabh
- Department of Animal Genetics and Breeding, Faculty of Veterinary and Animal Sciences, IAS, Banaras Hindu University, Varanasi, UP, India
| | - Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Sheikh Firdous Ahmad
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Rajat Varshney
- Bacteriology and Mycology Division, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Ankita Verma
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Naseer Ahmad Baba
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Satish Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Soni Kumari
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Anuj Chauhan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Pushpendra Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| | - Bharat Bhushan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, India
| |
Collapse
|
16
|
Jeon JH, Park DB, Woo SJ, Lee HR, Park OK, Park J, Rhie GE. Muramyl dipeptide potentiates a Bacillus anthracis poly-γ-d-glutamic acid capsule surrogate that induces maturation and activation of mouse dendritic cells. Cytokine 2018; 110:350-356. [PMID: 29656957 DOI: 10.1016/j.cyto.2018.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/28/2022]
Abstract
Poly-γ-d-glutamic acid (PGA) of anthrax is an important pathogenic factor due to its anti-phagocytic activity. Additionally, PGA has the ability to activate mouse macrophages for the secretion of cytokines through Toll-like receptor (TLR) 2. Peptidoglycan (PGN), a major bacterial cell-wall component, induces inflammatory responses in the host. We assessed whether PGA can induce maturation and cytokine expression in immature mouse dendritic cells (DCs) in the existence of muramyl dipeptide (MDP), the minimum motif of PGN with immunostimulatory activity. Stimulation of immature DCs with PGA or MDP alone augmented expression of costimulatory molecules and MHC class II proteins, which are all cell surface markers indicative of maturation. The observed effects were further enhanced by costimulation of PGA and MDP. PGA alone was sufficient to induce expression of TNF-α, IL-6, MCP-1, and MIP1-α, whereas MDP alone did not under the same conditions. Treatment with MDP enhanced PGA-induced expression of the tested inflammatory mediators; however, the synergistic effect found for PGA and MDP was not observed in TLR2- or nucleotide-binding oligomerization domain (NOD) 2-knockout DCs. Additionally, MDP augmented PGA-induced MAP kinases and NF-κB activation, which is crucial for expression of cytokines. Furthermore, MAP kinase and NF-κB inhibitors attenuated MDP enhancement of PGA-induced cytokine production. In addition, co-culture of splenocytes and PGA/MDP-matured DCs induced higher expression of IL-2 and IFN-γ compared to that of splenocytes and PGA-matured DCs. Collectively, our results suggest that PGA and MDP cooperatively induce inflammatory responses in mouse DCs through TLR2 and NOD2 via MAP kinase and NF-κB pathways, subsequently leading to lymphocyte activation.
Collapse
Affiliation(s)
- Jun Ho Jeon
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea
| | - Deok-Bum Park
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea
| | - Sun-Je Woo
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea
| | - Hae-Ri Lee
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea
| | - Ok-Kyu Park
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea
| | - Jungchan Park
- Department of Bioscience and Biotechnology, Hankuk University of Foreign Studies, Yongin 17035, Republic of Korea; Protein Research Center for Bioindustry, Hankuk University of Foreign Studies, Yongin 17035, Republic of Korea
| | - Gi-Eun Rhie
- Division of High-risk Pathogens, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Republic of Korea.
| |
Collapse
|
17
|
Bhattarai D, Worku T, Dad R, Rehman ZU, Gong X, Zhang S. Mechanism of pattern recognition receptors (PRRs) and host pathogen interplay in bovine mastitis. Microb Pathog 2018; 120:64-70. [PMID: 29635052 DOI: 10.1016/j.micpath.2018.04.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 04/01/2018] [Accepted: 04/06/2018] [Indexed: 02/08/2023]
Abstract
Bacterial infection in the mammary gland parenchyma induces local and subsequently systemic inflammation that results in a complex disease. Mastitis in bovine is the result of various factors which function together. This review is aimed to analyze the factors involved in the pathogenesis of common bacterial species for bovine mastitis. The bacterial growth patterns, signaling pathway and the pathogen-associated molecular patterns (PAMPs) which activate immune responses is discussed. Clear differences in bacterial infection pattern are shown between bacterial species and illustrated TLRs, NLRs and RLGs molecular mechanism for the initiation of intramammary infection. The underlying reasons for the differences and the resulting host response are analyzed. Understandings of the mechanisms that activate and regulate these responses are central to the development of efficient anticipatory and treatment management. The knowledge of bovine mammary gland to common mastitis causing pathogens with possible immune mechanism could be a new conceptual understanding for the prospect of mastitis control program.
Collapse
Affiliation(s)
- Dinesh Bhattarai
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430071, China.
| | - Tesfaye Worku
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430071, China
| | - Rahim Dad
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430071, China
| | - Zia Ur Rehman
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430071, China
| | - Xiaoling Gong
- The Agricultural Broadcasting and Television School in Hubei Province, Wuhan, 430064, China
| | - Shujun Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430071, China.
| |
Collapse
|
18
|
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: 5.5] [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
|
19
|
Rainard P, Foucras G, Fitzgerald JR, Watts JL, Koop G, Middleton JR. Knowledge gaps and research priorities in Staphylococcus aureus mastitis control. Transbound Emerg Dis 2017; 65 Suppl 1:149-165. [PMID: 28984427 DOI: 10.1111/tbed.12698] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Indexed: 12/31/2022]
Abstract
This study assessed knowledge gaps and suggested research priorities in the field of Staphylococcus aureus mastitis. Staphylococcus aureus infecting the mammary gland remains a major problem to the dairy industry worldwide because of its pathogenicity, contagiousness, persistence in the cow environment, colonization of skin or mucosal epithelia, and the poor curing efficacy of treatments. Staphylococcus aureus also constitutes a threat to public health due to food safety and antibiotic usage issues and the potential for bidirectional transmission of strains between humans and dairy animals (cows and small ruminants). Gaps have been identified in (i) understanding the molecular basis for pathogenesis of S. aureus mastitis, (ii) identifying staphylococcal antigens inducing protection and (iii) determining the cell-mediated immune responses to infection and vaccination. The recommended priorities for research are (i) improved diagnostic methods for early detection of infection and intervention through treatment or management, (ii) development of experimental models to investigate the strategies used by S. aureus to survive within the mammary gland and resist treatment with anti-microbials, (iii) investigation of the basis for cow-to-cow variation in response to S. aureus mastitis, (iv) identification of the immune responses (adaptive and innate) induced by infection or vaccination and (v) antibacterial discovery programmes to develop new, more effective, narrow spectrum antibacterial agents for the treatment of S. aureus mastitis. With the availability and ongoing improvement of molecular research tools, these objectives may not be out of reach in the future.
Collapse
Affiliation(s)
- P Rainard
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | - G Foucras
- IHAP, Université de Toulouse, INRA, ENVT, Toulouse, France
| | - J R Fitzgerald
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - J L Watts
- Zoetis, External Innovation-Anti-Infectives, VMRD, Kalamazoo, MI, USA
| | - G Koop
- Veterinary Medicine, University of Utrecht, Utrecht, The Netherlands
| | - J R Middleton
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| |
Collapse
|
20
|
Scumpia PO, Botten GA, Norman JS, Kelly-Scumpia KM, Spreafico R, Ruccia AR, Purbey PK, Thomas BJ, Modlin RL, Smale ST. Opposing roles of Toll-like receptor and cytosolic DNA-STING signaling pathways for Staphylococcus aureus cutaneous host defense. PLoS Pathog 2017; 13:e1006496. [PMID: 28704551 PMCID: PMC5526579 DOI: 10.1371/journal.ppat.1006496] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/25/2017] [Accepted: 06/27/2017] [Indexed: 01/01/2023] Open
Abstract
Successful host defense against pathogens requires innate immune recognition of the correct pathogen associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs) to trigger the appropriate gene program tailored to the pathogen. While many PRR pathways contribute to the innate immune response to specific pathogens, the relative importance of each pathway for the complete transcriptional program elicited has not been examined in detail. Herein, we used RNA-sequencing with wildtype and mutant macrophages to delineate the innate immune pathways contributing to the early transcriptional response to Staphylococcus aureus, a ubiquitous microorganism that can activate a wide variety of PRRs. Unexpectedly, two PRR pathways—the Toll-like receptor (TLR) and Stimulator of Interferon Gene (STING) pathways—were identified as dominant regulators of approximately 95% of the genes that were potently induced within the first four hours of macrophage infection with live S. aureus. TLR signaling predominantly activated a pro-inflammatory program while STING signaling activated an antiviral/type I interferon response with live but not killed S. aureus. This STING response was largely dependent on the cytosolic DNA sensor cyclic guanosine-adenosine synthase (cGAS). Using a cutaneous infection model, we found that the TLR and STING pathways played opposite roles in host defense to S. aureus. TLR signaling was required for host defense, with its absence reducing interleukin (IL)-1β production and neutrophil recruitment, resulting in increased bacterial growth. In contrast, absence of STING signaling had the opposite effect, enhancing the ability to restrict the infection. These results provide novel insights into the complex interplay of innate immune signaling pathways triggered by S. aureus and uncover opposing roles of TLR and STING in cutaneous host defense to S. aureus. Individual pathogen associated molecular patterns (PAMPs) induce gene expression in immune cells through distinct signaling pathways to protect cells from infection. However, pathogens typically possess many PAMPs, and the precise contribution of each PAMP to the gene expression program elicited by a live pathogen has not been clearly defined. Herein, we used gene expression profiling to examine the full early response of macrophages to Staphylococcus aureus, a major human opportunistic pathogen. Surprisingly, we found that two pathogen-sensing pathways, Toll-like receptor (TLR) and Stimulator of Interferon Signaling Gene (STING) pathways, contribute to the activation of ~95% of the genes induced by S. aureus infection. The remaining genes may be induced by hypoxia pathways. When the bacterium is dead, 98% of the gene induction occurs through TLR signaling, and neither STING nor hypoxia contributes greatly to the response. STING activation requires sensing of S. aureus DNA by the cytosolic DNA sensor, cGAS. During S. aureus skin infection, the TLR and STING pathways compete with each other to induce or suppress host defense, respectively, by counter regulating interleukin 1β production and neutrophil recruitment. A similar approach may allow delineation of the relative contributions of immune pathways in the response to various live pathogens.
Collapse
Affiliation(s)
- Philip O Scumpia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Giovanni A Botten
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Joshua S Norman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Kindra M Kelly-Scumpia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Roberto Spreafico
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America.,Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Amber R Ruccia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Prabhat K Purbey
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Brandon J Thomas
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Robert L Modlin
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America.,Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
21
|
Local immunization impacts the response of dairy cows to Escherichia coli mastitis. Sci Rep 2017; 7:3441. [PMID: 28611405 PMCID: PMC5469773 DOI: 10.1038/s41598-017-03724-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/03/2017] [Indexed: 01/13/2023] Open
Abstract
Current vaccines to Escherichia coli mastitis have shown some albeit limited efficacy. Their mode of action has not been documented, and immune responses protecting the mammary gland against E. coli are not completely understood. To improve our knowledge of mammary gland immune protection, cows immunized either intramuscularly or intramammarily with the E. coli P4 were submitted to a homologous mastitis challenge. A third group of mock-immunized cows serve as challenge controls. Local immunization modified favorably the course of infection, by improving bacterial clearance while limiting inflammation. Systemic clinical signs and reduction in milk secretion were also contained. This occurred with a modification of the cytokine profile, such as an increase in IFN-γ and a reduction in TNF-α concentrations in milk. Concentrations of IL-17A and IL-22 increased in milk at the onset of the inflammatory response and remained high up to the elimination of bacteria, but concentrations did not differ between groups. Accelerated bacteriological cure was not linked to an increase in the initial efficiency of phagocytosis in milk. Results support the idea that antibodies did not play a major role in the improvement, and that cell-mediated immunity is the key to understanding E. coli vaccine-induced protection of the mammary gland.
Collapse
|
22
|
17β-Estradiol and progesterone decrease MDP induced NOD2 expression in bovine mammary epithelial cells. Vet Immunol Immunopathol 2017; 188:59-64. [PMID: 28615128 DOI: 10.1016/j.vetimm.2017.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 11/20/2022]
Abstract
During the periparturient period, many neuroendocrine changes develop in cows. Periparturient hormone fluxes may adversely affect mammary gland immunity and mastitis susceptibility. 17β-Estradiol (E2) and progesterone (P4) have been reported to function on immune regulation, and their concentration fluctuates dramatically during the perinatal period. Nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) mediate numerous aspects of innate immunity in humans and experimental animals. This study aimed to explore the effects of E2 and P4 on NOD2 expression in bovine mammary epithelial cells (BMECs). BMECs were isolated and purified from bovine mammary tissue and treated with E2/P4 and muramyl dipeptide (MDP). After these treatments, the mRNA levels of NOD2, receptor-interacting protein kinase (RIP) 2, interleukin (IL) 1β, IL-6, IL-8 and tumor necrosis factor (TNF) α were assessed by quantitative real-time polymerase chain reaction (qRT-PCR) respectively, and the protein levels of NOD2 were analyzed by western blotting. The results showed that E2 and P4 decreased MDP-induced transcriptional expression of NOD2 and the downstream molecules. Moreover, E2 reduced MDP-induced NOD2 protein expression levels. Our study suggests that down-regulation of NOD2 by E2 and P4 may be one of the reasons for mastitis susceptibility in periparturient dairy cows.
Collapse
|
23
|
Rainard P. Mammary microbiota of dairy ruminants: fact or fiction? Vet Res 2017; 48:25. [PMID: 28412972 PMCID: PMC5392980 DOI: 10.1186/s13567-017-0429-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Explorations of how the complex microbial communities that inhabit different body sites might contribute to health and disease have prompted research on the ways the harmonious relationship between a host and its microbiota could be used to keep animals healthy in their production conditions. In particular, there is a growing interest in the bacterial signatures that can be found in the milk of healthy or mastitic dairy cows. The concept of sterility of the healthy mammary gland of dairy ruminants has been challenged by the results of studies using bacterial DNA-based methodology. The newly obtained data have led to the concept of the intramammary microbiota composed of a complex community of diverse bacteria. Accordingly, mammary gland infections are not mere infections by a bacterial pathogen, but the consequence of mammary dysbiosis. This article develops the logical implications of this paradigm shift and shows how this concept is incompatible with current knowledge concerning the innate and adaptive immune system of the mammary gland of dairy ruminants. It also highlights how the concept of mammary microbiota clashes with results of experimental infections induced under controlled conditions or large field experiments that demonstrated the efficacy of the current mastitis control measures.
Collapse
Affiliation(s)
- Pascal Rainard
- ISP, INRA, Université de Tours, UMR1282, 37380, Nouzilly, France.
| |
Collapse
|
24
|
Bulgari O, Dong X, Roca AL, Caroli AM, Loor JJ. Innate immune responses induced by lipopolysaccharide and lipoteichoic acid in primary goat mammary epithelial cells. J Anim Sci Biotechnol 2017; 8:29. [PMID: 28396748 PMCID: PMC5379715 DOI: 10.1186/s40104-017-0162-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/21/2017] [Indexed: 12/24/2022] Open
Abstract
Background Innate immune responses induced by in vitro stimulation of primary mammary epithelial cells (MEC) using Gram-negative lipopolysaccharide (LPS) and Gram-positive lipoteichoic acid (LTA) bacterial cell wall components are well- characterized in bovine species. The objective of the current study was to characterize the downstream regulation of the inflammatory response induced by Toll-like receptors in primary goat MEC (pgMEC). We performed quantitative real-time RT-PCR (qPCR) to measure mRNA levels of 9 genes involved in transcriptional regulation or antibacterial activity: Toll-like receptor 2 (TLR2), Toll-like receptor 4 (TLR4), prostaglandin-endoperoxide synthase 2 (PTGS2), interferon induced protein with tetratricopeptide repeats 3 (IFIT3), interferon regulatory factor 3 (IRF3), myeloid differentiation primary response 88 (MYD88), nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1), Toll interacting protein (TOLLIP), and lactoferrin (LTF). Furthermore, we analyzed 7 cytokines involved in Toll-like receptor signaling pathways: C-C motif chemokine ligand 2 (CCL2), C-C motif chemokine ligand 5 (CCL5), C-X-C motif chemokine ligand 6 (CXCL6), interleukin 8 (CXCL8), interleukin 1 beta (IL1B), interleukin 6 (IL6), and tumor necrosis factor alpha (TNF). Results Stimulation of pgMEC with LPS for 3 h led to an increase in expression of CCL2, CXCL6, IL6, CXCL8, PTGS2, IFIT3, MYD88, NFKB1, and TLR4 (P < 0.05). Except for IL6, and PTGS2, the same genes had greater expression than controls at 6 h post-LPS (P < 0.05). Expression of CCL5, PTGS2, IFIT3, NFKB1, TLR4, and TOLLIP was greater than controls after 3 h of incubation with LTA (P < 0.05). Compared to controls, stimulation with LTA for 6 h led to greater expression of PTGS2, IFIT3, NFKB1, and TOLLIP (P < 0.05) whereas the expression of CXCL6, CXCL8, and TLR4 was lower (P < 0.05). At 3 h incubation with both toxins compared to controls a greater expression (P < 0.05) of CCL2, CCL5, CXCL6, CXCL8, IL6, PTGS2, IFIT3, IRF3, MYD88, and NFKB1 was detected. After 6 h of incubation with both toxins, the expression of CCL2, CXCL6, IFIT3, MYD88, NFKB1, and TLR4 was higher than the controls (P < 0.05). Conclusions Data indicate that in the goat MEC, LTA induces a weaker inflammatory response than LPS. This may be related to the observation that gram-positive bacteria cause chronic mastitis more often than gram-negative infections. Electronic supplementary material The online version of this article (doi:10.1186/s40104-017-0162-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Omar Bulgari
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123 Italy
| | - Xianwen Dong
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA.,Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130 China
| | - Alfred L Roca
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Anna M Caroli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123 Italy
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| |
Collapse
|
25
|
Dhiman A, Mishra SK, Dubey PK, Goyal S, Sehgal M, Niranjan SK, Sodhi M, Mishra BP, Kataria RS. Identification of genetic variation in NOD-like receptor 2 gene and influence of polymorphism on gene structure and function in buffalo (Bubalus bubalis). Res Vet Sci 2017; 115:43-50. [PMID: 28135669 DOI: 10.1016/j.rvsc.2017.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/08/2017] [Accepted: 01/20/2017] [Indexed: 12/15/2022]
Abstract
Nucleotide-binding oligomerization domain (NOD)-like receptor 2 is one of the important mediators of innate as well as adaptive immune response to microbial infections. In this study, NOD-like receptor-2 was characterized by determining the full gene sequence and analyzing genetic diversity in Indian buffaloes. Sequence analysis of buffalo NOD2 revealed 3042 nucleotides long ORF, encoding 1013 amino acids from 12 exons. Domain structure analysis indicated existence of 8 leucine-rich repeat (LRR) domains in buffalo, cattle, sheep and mouse, along with central NACHT/NOD domain and two N-terminal CARD domains. Comparative sequence analysis among different buffalo breeds identified 46 polymorphic sites in NOD2 gene. Among coding region SNPs, 10 were non-synonymous, 7 synonymous and 3 were present in 5'UTR. Genotyping of two nsSNPs, revealed significant differences in the allele frequencies, distinguishing swamp and riverine buffaloes, having different utilities. Association analysis with mastitis in dairy buffaloes indicated significant variation in allelic frequencies at G1135A locus, between mastitis affected and non-affected animals. Further, NOD2 gene expression was quantified in different riverine buffalo tissues, using real-time PCR and lymph node displayed highest expression, compared to others organs included in the study. Overall, the study revealed buffalo NOD2 gene attributes, important to understand species specific immune response in ruminants.
Collapse
Affiliation(s)
- Asmita Dhiman
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - S K Mishra
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - P K Dubey
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - S Goyal
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - M Sehgal
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - S K Niranjan
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - M Sodhi
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - B P Mishra
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India
| | - R S Kataria
- ICAR-National Bureau of Animal Genetic Resources, GT Road By-Pass, Karnal, 132 001, Haryana, India.
| |
Collapse
|
26
|
Innate and Adaptive Immunity Synergize to Trigger Inflammation in the Mammary Gland. PLoS One 2016; 11:e0154172. [PMID: 27100324 PMCID: PMC4839698 DOI: 10.1371/journal.pone.0154172] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/08/2016] [Indexed: 12/21/2022] Open
Abstract
The mammary gland is able to detect and react to bacterial intrusion through innate immunity mechanisms, but mammary inflammation can also result from antigen-specific adaptive immunity. We postulated that innate and adaptive immune responses could synergize to trigger inflammation in the mammary gland. To test this hypothesis, we immunized cows with the model antigen ovalbumin and challenged the sensitized animals with either Escherichia coli lipopolysaccharide (LPS) as innate immunity agonist, ovalbumin as adaptive immunity agonist, or both agonists in three different udder quarters of lactating cows. There was a significant amplification of the initial milk leukocytosis in the quarters challenged with the two agonists compared to leukocytosis in quarters challenged with LPS or ovalbumin alone. This synergistic response occurred only with the cows that developed the ovalbumin-specific inflammatory response, and there were significant correlations between milk leukocytosis and production of IL-17A and IFN-γ in a whole-blood ovalbumin stimulation assay. The antigen-specific response induced substantial concentrations of IL-17A and IFN-γ in milk contrary to the response to LPS. Such a synergy at the onset of the reaction of the mammary gland suggests that induction of antigen-specific immune response with bacterial antigens could improve the initial immune response to infection, hence reducing the bacterial load and contributing to protection.
Collapse
|
27
|
Rainard P, Cunha P, Ledresseur M, Staub C, Touzé JL, Kempf F, Gilbert FB, Foucras G. Antigen-Specific Mammary Inflammation Depends on the Production of IL-17A and IFN-γ by Bovine CD4+ T Lymphocytes. PLoS One 2015; 10:e0137755. [PMID: 26375594 PMCID: PMC4573518 DOI: 10.1371/journal.pone.0137755] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/20/2015] [Indexed: 12/03/2022] Open
Abstract
Intramammary infusion of the antigen used to sensitize cows by the systemic route induces a local inflammation associated with neutrophil recruitment. We hypothesize that this form of delayed type hypersensitivity, which may occur naturally during infections or could be induced intentionally by vaccination, can impact the outcome of mammary gland infections. We immunized cows with ovalbumin to identify immunological correlates of antigen-specific mammary inflammation. Intraluminal injection of ovalbumin induced a mastitis characterized by a prompt tissue reaction (increase in teat wall thickness) and an intense influx of leukocytes into milk of 10 responder cows out of 14 immunized animals. The magnitude of the local inflammatory reaction, assessed through milk leukocytosis, correlated with antibody titers, skin thickness test, and production of IL-17A and IFN-γ in a whole-blood antigen stimulation assay (WBA). The production of these two cytokines significantly correlated with the magnitude of the milk leukocytosis following the ovalbumin intramammary challenge. The IL-17A and IFN-γ production in the WBA was dependent on the presence of CD4+ cells in blood samples. In vitro stimulation of peripheral blood lymphocytes with ovalbumin followed by stimulation with PMA/ionomycin allowed the identification by flow cytometry of CD4+ T cells producing either IL-17A, IFN-γ, or both cytokines. The results indicate that the antigen-specific WBA, and specifically IL-17A and IFN-γ production by circulating CD4+ cells, can be used as a predictor of mammary hypersensitivity to protein antigens. This prompts further studies aiming at determining how Th17 and/or Th1 lymphocytes modulate the immune response of the mammary gland to infection.
Collapse
Affiliation(s)
- Pascal Rainard
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
- * E-mail:
| | - Patricia Cunha
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Marion Ledresseur
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Christophe Staub
- UE1297, Unité Expérimentale de Physiologie Animale, UEPAO, INRA, Nouzilly, France
| | - Jean-Luc Touzé
- Université François Rabelais de Tours, Tours, France
- UMR0085, Physiologie de la Reproduction et du Comportement, INRA, Nouzilly, France
| | - Florent Kempf
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Florence B. Gilbert
- UMR1282, Infectiologie et Santé Publique, INRA, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
| | - Gilles Foucras
- INP, ENVT, Université de Toulouse, Toulouse, France
- UMR1225, Interactions Hôte Agents Pathogènes, INRA, Toulouse, France
| |
Collapse
|
28
|
Medina-Estrada I, Alva-Murillo N, López-Meza JE, Ochoa-Zarzosa A. Non-classical effects of prolactin on the innate immune response of bovine mammary epithelial cells: Implications during Staphylococcus aureus internalization. Microb Pathog 2015; 89:43-53. [PMID: 26341952 DOI: 10.1016/j.micpath.2015.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/24/2015] [Accepted: 08/24/2015] [Indexed: 11/17/2022]
Abstract
Staphylococcus aureus has the ability to invade mammary epithelial cells (bMECs) causing mastitis. This event depends primarily on the α5β1 integrin in the host cell. In addition, bMECs are a target for the hormone prolactin (PRL), which can regulate β1 integrin-dependent actions related to differentiation and lactation. Previously, we demonstrated that bovine PRL (bPRL, 5 ng/ml) stimulates S. aureus internalization into bMECs. TLR2 is important during S. aureus infections, but its activation by PRL has not yet been established. The objective of this study was to determine the role of α5β1 integrin and TLR2 during S. aureus internalization into bMECs stimulated with bPRL. We demonstrated that the prolactin-stimulated internalization of S. aureus decreases in response to the blockage of α5β1 integrin (∼ 80%) and TLR2 (∼ 80%). bPRL increases the membrane abundance (MA) of α5β1 integrin (∼ 20%) and induces TLR2 MA (∼ 2-fold). S. aureus reduces the α5β1 integrin MA in bMECs treated with bPRL (∼ 75%) but induces TLR2 MA in bMECs (∼ 3-fold). Bacteria and bPRL did not modify TLR2 MA compared with the hormone alone. S. aureus induces the activation of the transcription factor AP-1, which was inhibited in bMECs treated with bPRL and infected. In general, bPRL induces both pro- and anti-inflammatory responses in bMECs, which are abated in response to bacterial challenge. Interestingly, the canonical Stat-5 transcription factor was not activated in the challenged bMECs and/or treated with bPRL. Taken together, these results support novel functions of prolactin as a modulator of the innate immune response that do not involve the classical prolactin pathway.
Collapse
Affiliation(s)
- Ivan Medina-Estrada
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carretera Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico
| | - Nayeli Alva-Murillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carretera Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico
| | - Joel E López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carretera Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico
| | - Alejandra Ochoa-Zarzosa
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carretera Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico.
| |
Collapse
|
29
|
Bauer I, Günther J, Wheeler TT, Engelmann S, Seyfert HM. Extracellular milieu grossly alters pathogen-specific immune response of mammary epithelial cells. BMC Vet Res 2015. [PMID: 26219462 PMCID: PMC4518681 DOI: 10.1186/s12917-015-0489-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Considerably divergent data have been published from attempts to model the E. coli vs. S. aureus specific immune reaction of the udder using primary cultures of bovine mammary epithelial cells from cows (pbMEC). Some groups reported a swift, strong and transient inflammatory response against challenges with E. coli and only a weak and retarded response against S. aureus, in agreement with the respective reaction of the udder. Others found almost the reverse. Presence or absence of fetal calf serum distinguished the experimental setting between both groups. We examined here if this causes the divergent reaction of the pbMEC towards both pathogen species. We challenged pbMEC with proteins from heat killed E. coli or S. aureus pathogens or purified TLR2 and TLR4 ligands. The stimuli were applied in normal growth medium with (SM10) or without (SM0) 10% fetal calf serum, or in the basal medium supplemented with 10 mg/ml milk proteins (SM Milk). RESULTS Withdrawal of FCS slowed down and decreased the extent by which E. coli or LPS enhanced the expression of cyto- and chemokine encoding genes through impaired TLR4 signalling but enforced their expression during stimulation with S. aureus. SM Milk strongly quenched the induction of those genes. S. aureus strain specific differences in the reaction of the pbMEC could only be recorded in SM0. NF-κB factors were activated by E. coli in all stimulation media, but only to a small extent by S. aureus, solely in SM0. Purified ligands for TLR2 stimulated expression of those genes and activated NF-κB equally well in SM10 and SM0. The mRNA destabilizing factor tristetraproline was only induced by E. coli in SM10 and by purified PAMPs. CONCLUSIONS Our data cross validate the correctness of previously published divergent data on the pathogen-specific induction of key immune genes in pbMEC. The differences are due to the presence of FCS, modulating signalling through TLR4 and TLR-unrelated pathogen receptors. S. aureus does not substantially activate any TLR signalling in MEC. Rather, receptors distinct from TLRs perceive the presence of S. aureus and control the immune response against this pathogen in MEC.
Collapse
Affiliation(s)
- Isabel Bauer
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Juliane Günther
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Thomas T Wheeler
- Dairy Foods, AgResearch Ltd, Ruakura Research Centre, Hamilton, 3240, New Zealand.
| | - Susanne Engelmann
- Institute for Microbiology, Technical University of Braunschweig, Spielmannstraße 7, 38106, Braunschweig, Germany. .,Helmholtz Center for Infection Research, Microbial Proteomics, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| | - Hans-Martin Seyfert
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
30
|
Roussel P, Cunha P, Porcherie A, Petzl W, Gilbert FB, Riollet C, Zerbe H, Rainard P, Germon P. Investigating the contribution of IL-17A and IL-17F to the host response during Escherichia coli mastitis. Vet Res 2015; 46:56. [PMID: 26062913 PMCID: PMC4462179 DOI: 10.1186/s13567-015-0201-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/05/2015] [Indexed: 12/17/2022] Open
Abstract
Mastitis remains a major disease of cattle with a strong impact on the dairy industry. There is a growing interest in understanding how cell mediated immunity contributes to the defence of the mammary gland against invading mastitis causing bacteria. Cytokines belonging to the IL-17 family, and the cells that produce them, have been described as important modulators of the innate immunity, in particular that of epithelial cells. We report here that expression of IL-17A and IL-17F genes, encoding two members of the IL-17 family, are induced in udder tissues of cows experimentally infected with Escherichia coli. The impact of IL-17A on the innate response of bovine mammary epithelial cells was investigated using a newly isolated cell line, the PS cell line. We first showed that PS cells, similar to primary bovine mammary epithelial cells, were able to respond to agonists of TLR2 and to LPS, provided CD14 was added to the culture medium. We then showed that secretion of CXCL8 and transcription of innate immunity related-genes by PS cells were increased by IL-17A, in particular when these cells were stimulated with live E. coli bacteria. Together with data from the literature, these results support the hypothesis that IL-17A and IL-17 F could play an important role in mediating of host-pathogen interactions during mastitis.
Collapse
Affiliation(s)
- Perrine Roussel
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Patricia Cunha
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Adeline Porcherie
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Wolfram Petzl
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany.
| | - Florence B Gilbert
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Céline Riollet
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Holm Zerbe
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany.
| | - Pascal Rainard
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Pierre Germon
- INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| |
Collapse
|
31
|
Pereyra EAL, Dallard BE, Calvinho LF. [Aspects of the innate immune response to intramammary Staphylococcus aureus infections in cattle]. Rev Argent Microbiol 2015; 46:363-75. [PMID: 25576422 DOI: 10.1016/s0325-7541(14)70096-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 10/29/2014] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is the pathogen most frequently isolated from bovine mastitis worldwide, causing chronic intramammary infections that limit profitable dairying. The objective of this article is to characterize the mechanisms involved in S. aureus mammary gland infections considering two different aspects of the infectious process; on the one hand, the aspects involved in the host innate immune response and on the other hand, the capacity of this organism to evade the immune system and interact with different cell types. The exploration of S. aureus interactions with the immune response of bovine mammary gland will help identify targets to outline new preventive or curative alternatives for intramammary infections caused by this organism.
Collapse
Affiliation(s)
- Elizabet A L Pereyra
- Laboratorio de Biología Celular y Molecular Aplicada, Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Instituto de Ciencias Veterinarias del Litoral (ICIVET-Litoral), UNL-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Bibiana E Dallard
- Laboratorio de Biología Celular y Molecular Aplicada, Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Instituto de Ciencias Veterinarias del Litoral (ICIVET-Litoral), UNL-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Luis F Calvinho
- Estación Experimental Agropecuaria Rafaela, Instituto Nacional de Tecnología Agropecuaria (INTA), Santa Fe, Argentina; Facultad de Ciencias Veterinarias, UNL, Santa Fe, Argentina.
| |
Collapse
|
32
|
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
|
33
|
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: 85] [Impact Index Per Article: 7.7] [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
|
34
|
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: 1.8] [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
|
35
|
Lind M, Sipka AS, Schuberth HJ, Blutke A, Wanke R, Sauter-Louis C, Duda KA, Holst O, Rainard P, Germon P, Zerbe H, Petzl W. Location-specific expression of chemokines, TNF-α and S100 proteins in a teat explant model. Innate Immun 2014; 21:322-31. [DOI: 10.1177/1753425914539820] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The distal compartments of the udder are the first to interact with invading pathogens. The regulatory and effector functions of two major teat regions [Fürstenberg's rosette (FR); teat cistern (TC)] are largely unknown. The objective of this study was to establish an in vitro model with explants of the FR and the TC to analyse their response towards Escherichia coli LPS and Staphylococcus aureus lipoteichoic acid (LTA). Quantitative stereological analysis confirmed differences in the cellular composition of FR and TC explants. Chemokine (CXCL8, CCL5, CCL20) and TNF-α mRNA were expressed at low levels in both locations. Explant stimulation with LPS increased the mRNA abundance of all tested chemokines and TNF-α. Stimulation with LTA only induced CCL20 and CXCL8. LPS- and LTA-stimulated explant supernatants contained CXCL8 and CXCL3. Supernatants significantly attracted neutrophils in vitro. Compared with TC, the FR showed high constitutive mRNA expression of S100 proteins (A8, A9, A12). In the TC, both LPS and LTA significantly induced S100A8, whereas S100A9 and S100A12 expression was only induced by LPS. The novel model system underpins the role of the teat for recognising pathogens and shaping a pathogen- and location-specific immune response.
Collapse
Affiliation(s)
- Monique Lind
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| | - Anja S Sipka
- Department for Population Medicine and Diagnostic Services, Cornell University, Ithaca, NY, USA
| | | | - Andreas Blutke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| | - Carola Sauter-Louis
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| | - Katarzyna A Duda
- Division of Structural Biochemistry, Research Centre Borstel, Leibniz-Centre for Medicine and Biosciences, Airway Research Center North (ARCN); German Centre for Lung Research (DZL), Borstel, Germany
| | - Otto Holst
- Division of Structural Biochemistry, Research Centre Borstel, Leibniz-Centre for Medicine and Biosciences, Airway Research Center North (ARCN); German Centre for Lung Research (DZL), Borstel, Germany
| | - Pascal Rainard
- INRA, UMR1282 Infectiologie et Santé Publique (ISP), Nouzilly, France
| | - Pierre Germon
- INRA, UMR1282 Infectiologie et Santé Publique (ISP), Nouzilly, France
| | - Holm Zerbe
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| | - Wolfram Petzl
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians University Munich, Oberschleissheim, Germany
| |
Collapse
|
36
|
Hu Q, Cui X, Tao L, Xiu L, Wang T, Wang X. Staphylococcus aureusInduces Apoptosis in Primary Bovine Mammary Epithelial Cells Through Fas-FADD Death Receptor-Linked Caspase-8 Signaling. DNA Cell Biol 2014; 33:388-97. [DOI: 10.1089/dna.2013.2195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Qingliang Hu
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xinjie Cui
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Lin Tao
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Lei Xiu
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Ting Wang
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xiao Wang
- College of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| |
Collapse
|
37
|
Liu M, Song S, Li H, Jiang X, Yin P, Wan C, Liu X, Liu F, Xu J. The protective effect of caffeic acid against inflammation injury of primary bovine mammary epithelial cells induced by lipopolysaccharide. J Dairy Sci 2014; 97:2856-65. [PMID: 24612802 DOI: 10.3168/jds.2013-7600] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/21/2014] [Indexed: 01/03/2023]
Abstract
Caffeic acid possesses multiple biological effects, such as antibacterial, antioxidant, antiinflammatory, and anticancer growth; however, what effects it has on bovine mastitis have not been investigated. The aim of this study was to verify the antiinflammatory properties of caffeic acid on the inflammatory response of primary bovine mammary epithelial cells (bMEC) induced by lipopolysaccharide (LPS), and to clarify the possible underlying mechanism. Bovine mammary epithelial cells were treated with various concentrations (10, 50, 100, and 200 μg/mL) of LPS for 3, 6, 12, and 18 h; the results showed that LPS significantly inhibited cell viability in a time- and dose-dependent manner. When cells were treated with LPS (50 μg/mL) for 12h, the cell membrane permeability significantly increased, which promoted cell apoptosis. Various concentrations (10, 25, and 50 μg/mL) of caffeic acid could weaken the inflammation injury of bMEC induced by LPS without cytotoxicity. Proinflammatory cytokines (IL-8, IL-1β, IL-6, and tumor necrosis factor α) from bMEC were decreased. Nuclear transcription factor κB activity was weakened via blocking κB inhibitor α degradation and p65 phosphorylation. All these showed that the protective effect of caffeic acid on LPS-induced inflammation injury in bMEC was at least partly achieved by the decreased production of proinflammatory cytokines mediated by the effect of reducing the κB inhibitor α degradation and p65 phosphorylation in the nuclear transcription factor κB pathway. The use of caffeic acid would be beneficial in dairy cows during Escherichia coli mastitis as a safe and natural antiinflammatory drug.
Collapse
Affiliation(s)
- Mingjiang Liu
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Shixiu Song
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Huanrong Li
- College of Animal Science and Technology, Beijing University of Agriculture (BUA), Beijing 102206, P. R. China
| | - Xiaoyu Jiang
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Peng Yin
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Changrong Wan
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Xiaoxi Liu
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China
| | - Fenghua Liu
- College of Animal Science and Technology, Beijing University of Agriculture (BUA), Beijing 102206, P. R. China.
| | - Jianqin Xu
- College of Veterinary Medicine, China Agricultural University (CAU), Beijing 100193, P. R. China.
| |
Collapse
|
38
|
Ahn KB, Jeon JH, Baik JE, Park OJ, Kang SS, Yun CH, Park JH, Han SH. Muramyl dipeptide potentiates staphylococcal lipoteichoic acid induction of cyclooxygenase-2 expression in macrophages. Microbes Infect 2014; 16:153-60. [DOI: 10.1016/j.micinf.2013.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 10/01/2013] [Accepted: 10/25/2013] [Indexed: 01/01/2023]
|
39
|
Lewandowska-Sabat AM, Boman GM, Downing A, Talbot R, Storset AK, Olsaker I. The early phase transcriptome of bovine monocyte-derived macrophages infected with Staphylococcus aureus in vitro. BMC Genomics 2013; 14:891. [PMID: 24341851 PMCID: PMC3878444 DOI: 10.1186/1471-2164-14-891] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 12/03/2013] [Indexed: 12/18/2022] Open
Abstract
Background In the mammary gland, local recruitment and action of macrophages is a key immunological defence mechanism against infection. Macrophages are members of the innate immune system, serve as the first line of the defence against invading pathogens and are critical effectors and regulators of inflammation. We have examined the early phase response of bovine macrophages to infection with live Staphylococcus aureus. Genome-wide transcript profiling of blood monocyte-derived macrophages from six Norwegian Red heifers infected with live S. aureus for 2 and 6 hours in vitro was performed. Results About 420 of the 17 000 genes on the ARK-Genomics bovine cDNA array were differentially regulated at 6 hours post infection. Approximately 70% of the responding genes had a known identity (Entrez Gene ID) and were used in the identification of overrepresented pathways and biological functions in the dataset. Analysis of a subset of differentially regulated genes (List eQG) obtained by comparison with data from genome-wide association mapping in Norwegian Red cattle identified anti-inflammatory cytokines interleukin 4 and interleukin 13 as putative expression quantitative trait loci, suggesting that S. aureus infection triggers alternative activation of macrophages. Moreover, several classical activation pathways were found, mainly cellular immune response and cytokine signaling pathways, i.e. triggering receptor expressed on myeloid cells 1 (TREM1) and nucleotide-binding and oligomerization domain-like receptor (NLR) pathways. Tumor necrosis factor receptor superfamily member 5 (CD40 ligand) was identified as an upstream regulator which points toward CD40 likely acting as a co-stimulatory receptor during Toll-like receptor 2(TLR2)-mediated inflammatory response of bovine macrophages to S. aureus infection. Furthermore, peptidoglycan was identified as an upstream regulator in the List eQG, which indicates that this bacterial cell-wall component might be pivotal in macrophage intracellular bacterial recognition during early inflammation. Conclusions Here we have shown that in vitro infection of bovine macrophages with live S. aureus induced both alternative and classical activation pathways. Alternative activation of macrophages may be a mechanism contributing to intracellular persistence of S. aureus in the course of inflammation such as during mastitis in dairy cattle.
Collapse
Affiliation(s)
| | | | | | | | | | - Ingrid Olsaker
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, P,O, Box 8146 Dep, NO-0033 Oslo, Norway.
| |
Collapse
|
40
|
Konsavage WM, Roper JN, Ishmael FT, Yochum GS. The Myc 3' Wnt responsive element regulates neutrophil recruitment after acute colonic injury in mice. Dig Dis Sci 2013; 58:2858-2867. [PMID: 23640071 PMCID: PMC4104363 DOI: 10.1007/s10620-013-2686-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 04/09/2013] [Indexed: 01/24/2023]
Abstract
BACKGROUND The Wnt/β-catenin pathway regulates intestinal development, homeostasis, and regeneration after injury. Wnt/β-catenin signaling drives intestinal proliferation by activating expression of the c-Myc proto-oncogene (Myc) through the Myc 3' Wnt responsive DNA element (Myc 3' WRE). In a previous study, we found that deletion of the Myc 3' WRE in mice caused increased MYC expression and increased cellular proliferation in the colon. When damaged by dextran sodium sulfate (DSS), the increased proliferative capacity of Myc 3' WRE(-/-) colonocytes resulted in a more rapid recovery compared with wild-type (WT) mice. In that study, we did not examine involvement of the immune system in colonic regeneration. PURPOSE To characterize the innate immune response in Myc 3' WRE(-/-) and WT mice during and after DSS-induced colonic injury. METHODS Mice were fed 2.5 % DSS in their drinking water for five days to induce colonic damage and were then returned to normal water for two or four days to recover. Colonic sections were prepared and neutrophils and macrophages were analyzed by immunohistochemistry. Cytokine and chemokine levels were analyzed by probing a cytokine array with colonic lysates. RESULTS In comparison with WT mice, there was enhanced leukocyte infiltration into the colonic mucosal and submucosal layers of Myc 3' WRE(-/-) mice after DSS damage. Levels of activated neutrophils were substantially increased in damaged Myc 3' WRE(-/-) colons as were levels of the neutrophil chemoattractants C5/C5a, CXCL1, and CXCL2. CONCLUSION The Myc 3' WRE regulates neutrophil infiltration into DSS-damaged colons.
Collapse
Affiliation(s)
- Wesley M. Konsavage
- The Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Dr. H171, Hershey, PA 17033, USA
| | - Jennifer N. Roper
- The Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Dr. H171, Hershey, PA 17033, USA
| | - Faoud T. Ishmael
- The Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Dr. H171, Hershey, PA 17033, USA
| | - Gregory S. Yochum
- The Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Dr. H171, Hershey, PA 17033, USA
| |
Collapse
|
41
|
Gilbert FB, Cunha P, Jensen K, Glass EJ, Foucras G, Robert-Granié C, Rupp R, Rainard P. Differential response of bovine mammary epithelial cells to Staphylococcus aureus or Escherichia coli agonists of the innate immune system. Vet Res 2013; 44:40. [PMID: 23758654 PMCID: PMC3686618 DOI: 10.1186/1297-9716-44-40] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 05/17/2013] [Indexed: 12/03/2022] Open
Abstract
Mastitis caused by Escherichia coli and Staphylococcus aureus is a major pathology of dairy cows. To better understand the differential response of the mammary gland to these two pathogens, we stimulated bovine mammary epithelial cells (bMEC) with either E. coli crude lipopolysaccharide (LPS) or with S. aureus culture supernatant (SaS) to compare the transcriptomic profiles of the initial bMEC response. By using HEK 293 reporter cells for pattern recognition receptors, the LPS preparation was found to stimulate TLR2 and TLR4 but not TLR5, Nod1 or Nod2, whereas SaS stimulated TLR2. Biochemical analysis revealed that lipoteichoic acid, protein A and α-hemolysin were all present in SaS, and bMEC were found to be responsive to each of these molecules. Transcriptome profiling revealed a core innate immune response partly shared by LPS and SaS. However, LPS induced expression of a significant higher number of genes and the fold changes were of greater magnitude than those induced by SaS. Microarray data analysis suggests that the activation pathways and the early chemokine and cytokine production preceded the defense and stress responses. A major differential response was the activation of the type I IFN pathway by LPS but not by SaS. The higher upregulation of chemokines (Cxcl10, Ccl2, Ccl5 and Ccl20) that target mononuclear leucocytes by LPS than by SaS is likely to be related to the differential activation of the type I IFN pathway, and could induce a different profile of the initial recruitment of leucocytes. The MEC responses to the two stimuli were different, as LPS was associated with NF-κB and Fas signaling pathways, whereas SaS was associated with AP-1 and IL-17A signaling pathways. It is noteworthy that at the protein level secretion of TNF-α and IL-1β was not induced by either stimulus. These results suggest that the response of MEC to diffusible stimuli from E. coli and S. aureus contributes to the onset of the response with differential leucocyte recruitment and distinct inflammatory and innate immune reactions of the mammary gland to infection.
Collapse
|
42
|
Lewis D, Chan D, Pinheiro D, Armitage‐Chan E, Garden O. The immunopathology of sepsis: pathogen recognition, systemic inflammation, the compensatory anti-inflammatory response, and regulatory T cells. J Vet Intern Med 2012; 26:457-82. [PMID: 22428780 PMCID: PMC7166777 DOI: 10.1111/j.1939-1676.2012.00905.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 11/28/2011] [Accepted: 02/07/2012] [Indexed: 02/06/2023] Open
Abstract
Sepsis, the systemic inflammatory response to infection, represents the major cause of death in critically ill veterinary patients. Whereas important advances in our understanding of the pathophysiology of this syndrome have been made, much remains to be elucidated. There is general agreement on the key interaction between pathogen-associated molecular patterns and cells of the innate immune system, and the amplification of the host response generated by pro-inflammatory cytokines. More recently, the concept of immunoparalysis in sepsis has also been advanced, together with an increasing recognition of the interplay between regulatory T cells and the innate immune response. However, the heterogeneous nature of this syndrome and the difficulty of modeling it in vitro or in vivo has both frustrated the advancement of new therapies and emphasized the continuing importance of patient-based clinical research in this area of human and veterinary medicine.
Collapse
Affiliation(s)
- D.H. Lewis
- Department of Veterinary Clinical SciencesThe Royal Veterinary CollegeHatfield CampusHertfordshire,UK (Lewis, Chan, Garden)
- Present address:
Langford Veterinary ServicesSmall Animal HospitalLangford HouseLangfordBristol, BS40 5DUUK
| | - D.L. Chan
- Department of Veterinary Clinical SciencesThe Royal Veterinary CollegeHatfield CampusHertfordshire,UK (Lewis, Chan, Garden)
| | - D. Pinheiro
- Regulatory T Cell LaboratoryThe Royal Veterinary CollegeCamden Campus, LondonNW1 OTUUK (Pinheiro, Garden)
| | - E. Armitage‐Chan
- Davies Veterinary SpecialistsManor Farm Business ParkHertfordshireSG5 3HR, UK (Armitage‐Chan)
| | - O.A. Garden
- Department of Veterinary Clinical SciencesThe Royal Veterinary CollegeHatfield CampusHertfordshire,UK (Lewis, Chan, Garden)
- Regulatory T Cell LaboratoryThe Royal Veterinary CollegeCamden Campus, LondonNW1 OTUUK (Pinheiro, Garden)
| |
Collapse
|
43
|
Kim H, Jung BJ, Jung JH, Kim JY, Chung SK, Chung DK. Lactobacillus plantarum lipoteichoic acid alleviates TNF-α-induced inflammation in the HT-29 intestinal epithelial cell line. Mol Cells 2012; 33:479-86. [PMID: 22526394 PMCID: PMC3887727 DOI: 10.1007/s10059-012-2266-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 03/08/2012] [Accepted: 03/08/2012] [Indexed: 01/04/2023] Open
Abstract
We recently observed that lipoteichoic acid (LTA) isolated from Lactobacillus plantarum inhibited endotoxin-mediated inflammation of the immune cells and septic shock in a mouse model. Here, we examined the inhibitory role of L. plantarum LTA (pLTA) on the inflammatory responses of intestinal epithelial cells (IEC). The human colon cell line, HT-29, increased interleukin (IL)-8 expression in response to recombinant human tumor necrosis factor (TNF)-alpha, but not in response to bacterial ligands and interferon (IFN)-gamma. TNF-α also increased the production of inducible nitric oxide synthase (iNOS), nitric oxide (NO), and intercellular adhesion molecule 1 (ICAM-1) through activation of p38 mitogen-activated protein kinase (MAPK) from HT-29 cells. However, the inflammatory response of HT-29 on TNF-α stimulation was significantly inhibited by pLTA treatment. This pLTA-mediated inhibition accompanied the inhibition of nuclear factor (NF)-kappa B and MAPKs. Our data suggest that pLTA regulates cytokine-mediated immune responses and may be a good candidate for maintaining intestinal homeostasis against excessive inflammation.
Collapse
Affiliation(s)
- Hangeun Kim
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104,
USA
| | - Bong Jun Jung
- School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin 449-701,
Korea
| | - Ji Hae Jung
- School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin 449-701,
Korea
| | - Joo Yun Kim
- School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin 449-701,
Korea
| | - Sung Kyun Chung
- Department of Dental Hygiene, Shinheung College, Uijeongbu 480-701,
Korea
| | - Dae Kyun Chung
- School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin 449-701,
Korea
- Skin Biotechnology Center, Kyung Hee University, Yongin 449-701,
Korea
- RNA Inc., College of Life Science, Kyung Hee University, Yongin 449-701,
Korea
| |
Collapse
|
44
|
The innate immune response of the bovine mammary gland to bacterial infection. Vet J 2012; 192:148-52. [PMID: 22498784 DOI: 10.1016/j.tvjl.2011.09.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 09/16/2011] [Accepted: 09/21/2011] [Indexed: 01/18/2023]
Abstract
Intra-mammary (IM) bacterial infection in cattle can result in clinical outcomes that range from being acute and life-threatening to those that are chronic and sub-clinical. The typical bacteria involved in IM bacterial infections activate the mammary immune system in different ways which can influence the severity of the outcome. A clear understanding of the mechanisms that activate and regulate this response is central to the development of effective preventative and treatment regimes. This review focuses on the different immune responses of the bovine mammary gland to common mastitis-causing pathogens. There is special emphasis on comparing the responses to Escherichia coli and Staphylococcus aureus infections, as these are typically associated, respectively, with acute/severe and chronic/sub-clinical forms of the disease.
Collapse
|
45
|
Bonnefont CMD, Rainard P, Cunha P, Gilbert FB, Toufeer M, Aurel MR, Rupp R, Foucras G. Genetic susceptibility to S. aureus mastitis in sheep: differential expression of mammary epithelial cells in response to live bacteria or supernatant. Physiol Genomics 2012; 44:403-16. [PMID: 22337903 DOI: 10.1152/physiolgenomics.00155.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Staphylococcus aureus is a prevalent pathogen for mastitis in dairy ruminants and is responsible for both clinical and subclinical mastitis. Mammary epithelial cells (MEC) represent not only a physical barrier against bacterial invasion but are also active players of the innate immune response permitting infection clearance. To decipher their functions in general and in animals showing different levels of genetic predisposition to Staphylococcus in particular, MEC from ewes undergoing a divergent selection on milk somatic cell count were stimulated by S. aureus. MEC response was also studied according to the stimulation condition with live bacteria or culture supernatant. The early MEC response was studied during a 5 h time course by microarray to identify differentially expressed genes with regard to the host genetic background and as a function of the conditions of stimulation. In both conditions of stimulation, metabolic processes were altered, the apoptosis-associated pathways were considerably modified, and inflammatory and immune responses were enhanced with the upregulation of il1a, il1b, and tnfa and several chemokines known to enhance neutrophil (cxcl8) or mononuclear leukocyte (ccl20) recruitment. Genes associated with oxidative stress were increased after live bacteria stimulation, whereas immune response-related genes were higher after supernatant stimulation in the early phase. Only 20 genes were differentially expressed between Staphylococcus spp-mastitis resistant and susceptible animals without any clearly defined role on the control of infection. To conclude, this suggests that MEC may not represent the cell type at the origin of the difference of mastitis susceptibility, at least as demonstrated in our genetic model. Supernatant or heat-killed S. aureus produce biological effects that are essentially different from those induced by live bacteria.
Collapse
Affiliation(s)
- Cécile M D Bonnefont
- Université de Toulouse, Institut National Polytechnique (INP), École Nationale Vétérinaire de Toulouse (ENVT), Unité Mixte de Recherche (UMR)1225, Interactions Hôtes - Agents Pathogènes (IHAP), Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Porcherie A, Cunha P, Trotereau A, Roussel P, Gilbert FB, Rainard P, Germon P. Repertoire of Escherichia coli agonists sensed by innate immunity receptors of the bovine udder and mammary epithelial cells. Vet Res 2012; 43:14. [PMID: 22330199 PMCID: PMC3305352 DOI: 10.1186/1297-9716-43-14] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 02/13/2012] [Indexed: 01/01/2023] Open
Abstract
Escherichia coli is a frequent cause of clinical mastitis in dairy cows. It has been shown that a prompt response of the mammary gland after E. coli entry into the lumen of the gland is required to control the infection, which means that the early detection of bacteria is of prime importance. Yet, apart from lipopolysaccharide (LPS), little is known of the bacterial components which are detected by the mammary innate immune system. We investigated the repertoire of potential bacterial agonists sensed by the udder and bovine mammary epithelial cells (bMEC) during E. coli mastitis by using purified or synthetic molecular surrogates of bacterial agonists of identified pattern-recognition receptors (PRRs). The production of CXCL8 and the influx of leucocytes in milk were the readouts of reactivity of stimulated cultured bMEC and challenged udders, respectively. Quantitative PCR revealed that bMEC in culture expressed the nucleotide oligomerization domain receptors NOD1 and NOD2, along with the Toll-like receptors TLR1, TLR2, TLR4, and TLR6, but hardly TLR5. In line with expression data, bMEC proved to react to the cognate agonists C12-iE-DAP (NOD1), Pam3CSK4 (TLR1/2), Pam2CSK4 (TLR2/6), pure LPS (TLR4), but not to flagellin (TLR5). As the udder reactivity to NOD1 and TLR5 agonists has never been reported, we tested whether the mammary gland reacted to intramammary infusion of C12-iE-DAP or flagellin. The udder reacted to C12-iE-DAP, but not to flagellin, in line with the reactivity of bMEC. These results extend our knowledge of the reactivity of the bovine mammary gland to bacterial agonists of the innate immune system, and suggest that E. coli can be recognized by several PRRs including NOD1, but unexpectedly not by TLR5. The way the mammary gland senses E. coli is likely to shape the innate immune response and finally the outcome of E. coli mastitis.
Collapse
Affiliation(s)
- Adeline Porcherie
- INRA, UMR 1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Initial recognition of bacteria by the innate immune system is thought to occur primarily by germline-encoded pattern recognition receptors (PRRs). These receptors are present in multiple compartments of host cells and are thus capable of surveying both the intracellular and extracellular milieu for bacteria. It has generally been presumed that the cellular location of these receptors dictates what type of bacteria they respond to: extracellular bacteria being recognized by cell surface receptors, such as certain Toll-like receptors, and bacteria that are capable of breaching the plasma membrane and entering the cytoplasm, being sensed by cytoplasmic receptors, including the Nod-like receptors (NLRs). Increasingly, it is becoming apparent that this is a false dichotomy and that extracellular bacteria can be sensed by cytoplasmic PRRs and this is crucial for controlling the levels of these bacteria. In this review, we discuss the role of two NLRs, Nod1 and Nod2, in the recognition of and response to extracellular bacteria.
Collapse
Affiliation(s)
- Thomas B Clarke
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6076, USA
| | | |
Collapse
|
48
|
Aitken SL, Corl CM, Sordillo LM. Immunopathology of mastitis: insights into disease recognition and resolution. J Mammary Gland Biol Neoplasia 2011; 16:291-304. [PMID: 21938490 DOI: 10.1007/s10911-011-9230-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/01/2011] [Indexed: 01/02/2023] Open
Abstract
Mastitis is an inflammation of the mammary gland commonly caused by bacterial infection. The inflammatory process is a normal and necessary immunological response to invading pathogens. The purpose of host inflammatory responses is to eliminate the source of tissue injury, restore immune homeostasis, and return tissues to normal function. The inflammatory cascade results not only in the escalation of local antimicrobial factors, but also in the increased movement of leukocytes and plasma components from the blood that may cause damage to host tissues. A precarious balance between pro-inflammatory and pro-resolving mechanisms is needed to ensure optimal bacterial clearance and the prompt return to immune homeostasis. Therefore, inflammatory responses must be tightly regulated to avoid bystander damage to the milk synthesizing tissues of the mammary gland. The defense mechanisms of the mammary gland function optimally when invading bacteria are recognized promptly, the initial inflammatory response is adequate to rapidly eliminate the infection, and the mammary gland is returned to normal function quickly without any noticeable clinical symptoms. Suboptimal or dysfunctional mammary gland defenses, however, may contribute to the development of severe acute inflammation or chronic mastitis that adversely affects the quantity and quality of milk. This review will summarize critical mammary gland defense mechanisms that are necessary for immune surveillance and the rapid elimination of mastitis-causing organisms. Situations in which diminished efficiency of innate or adaptive mammary gland immune responses may contribute to disease pathogenesis will also be discussed. A better understanding of the complex interactions between mammary gland defenses and mastitis-causing pathogens should prove useful for the future control of intramammary infections.
Collapse
Affiliation(s)
- Stacey L Aitken
- G300 Veterinary Medical Center, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | | | | |
Collapse
|
49
|
Bougarn S, Cunha P, Gilbert FB, Harmache A, Foucras G, Rainard P. Staphylococcal-associated molecular patterns enhance expression of immune defense genes induced by IL-17 in mammary epithelial cells. Cytokine 2011; 56:749-59. [PMID: 22004923 DOI: 10.1016/j.cyto.2011.09.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 07/12/2011] [Accepted: 09/22/2011] [Indexed: 12/14/2022]
Abstract
Interleukin-17A (IL-17A) and IL-17F have been shown to mediate a crucial crosstalk between the immune system and various epithelial tissues, stimulating various defensive mechanisms to bacterial infections. A number of studies have characterized the response to IL-17A and IL-17F of epithelial cells from airways, intestine, and skin, but not from the mammary gland. To evaluate the potential contribution of IL-17 to the immune defense of the mammary gland, we analyzed the effects of recombinant bovine IL-17A and IL-17F on primary bovine mammary epithelial cells (MEC) by quantitative PCR and ELISA. We found expression (mRNA) of the two components of the IL-17 receptor complex, IL-17RA and IL-17RC, in mammary tissue and MEC in vitro. The expression of a number of genes encoding cytokines, chemokines and proteins endowed with antibacterial activities was increased by IL-17A, and to a lesser extent by IL-17F, but the magnitude of responses was modest. As expected, responses were augmented by the combination of IL-17A or IL-17F with TNF-α. Interestingly, responses of a few of the tested genes, such as IL8, CCL20, iNOS, and CfB, were augmented by the combination of IL-17A with staphylococcal lipoteichoic acid or muramyl dipeptide, bacterial agonists of the innate immune system. This can be interpreted as indicating that IL-17A and IL-17F are tailored to exert their full potential in a septic environment. MEC responses were characterized by the expression of chemokines targeting not only neutrophils (CXCL3 and CXCL8) but also mononuclear leucocytes (CCL2, CCL20). Production of IL-6 was low and the inflammatory cytokines TNF-α and IL-1β were expressed (mRNA) but proteins were not secreted. Altogether, our results suggest that IL-17A and IL-17F have a potential to modulate the mammary gland immune response to mastitis-causing pathogens.
Collapse
Affiliation(s)
- Salim Bougarn
- INRA, UR1282 Infectiologie Animale et Santé Publique (IASP), F-37380 Nouzilly, France
| | | | | | | | | | | |
Collapse
|
50
|
Fromageau A, Cunha P, Gilbert FB, Rainard P. Purified Staphylococcus aureus leukotoxin LukM/F' does not trigger inflammation in the bovine mammary gland. Microb Pathog 2011; 51:396-401. [PMID: 21951578 DOI: 10.1016/j.micpath.2011.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 09/12/2011] [Accepted: 09/15/2011] [Indexed: 12/01/2022]
Abstract
An early recruitment of neutrophils in mammary tissue and milk is considered an important component of the defense of the mammary gland against Staphylococcus aureus. We investigated whether the leukotoxin LukM/F', which is produced by a proportion of mastitis-causing strains of S. aureus, would be able to trigger inflammation in the udder. Infusion of purified LukM/F' toxin in lactating mammary glands did not cause neutrophil influx in milk, showing that the toxin was not able to cause mastitis on its own. Purified LukM/F' did not kill or stimulate mammary epithelial cells in culture. As expected, LukM bound to mammary macrophages and the complete LukM/F' toxin killed these cells, but subcytotoxic LukM/F' concentrations did not induce secretion of IL-8, TNF-α, IL-1β or IL-6 by macrophages. On the contrary, the production of these pro-inflammatory mediators by adhesion-stimulated macrophages was reduced. Overall, these results indicate that purified leukotoxin LukM/F' is not likely to contribute to the initiation of the inflammatory response and could even play an anti-inflammatory role in the mammary gland by inactivating macrophages.
Collapse
Affiliation(s)
- Angélina Fromageau
- INRA, UR 1282 Infectiologie Animale et Santé Publique, IASP Bat. 311, F-37380 Nouzilly, France
| | | | | | | |
Collapse
|