1
|
Mavangira V. Immunology of the Bovine Mammary Gland: Advances in Recent Years. Vet Clin North Am Food Anim Pract 2025; 41:137-154. [PMID: 40274420 DOI: 10.1016/j.cvfa.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Bovine mastitis remains the most costly disease in the dairy industry due to its impact on milk production, milk quality, and animal welfare. Optimal immunity is essential for the mammary gland to resist infections, efficiently clear them, and limit damage to mammary tissue. This article highlights recent advancements built on previous knowledge exploring opportunities for enhancing mammary gland immune responses. Ultimately, these improvements aim to increase the resistance of the mammary gland to infections and reduce the costs associated with this disease in the dairy industry.
Collapse
Affiliation(s)
- Vengai Mavangira
- Veterinary Diagnostic and Production Animal Medicine Department, Iowa State University, 1809 South Riverside Road, Ames, IA 50011, USA.
| |
Collapse
|
2
|
Burucúa MM, Risalde MA, Langellotti CA, Quintana S, Odeón AC, Cobo ER, Cutrera AP, Pérez SE, Marin MS. Immunoregulatory and antiviral effect mediated by TLR7 and BMAP28 interaction in bovine alphaherpesvirus-infected respiratory primary cultures. Vet Microbiol 2025; 300:110342. [PMID: 39705819 DOI: 10.1016/j.vetmic.2024.110342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Varicellovirus bovinealpha 1 and 5 (formerly bovine alphaherpesvirus type 1 and 5, BoAHV-1 and BoAHV-5) are closely related and can be isolated from similar clinical conditions, including respiratory and nervous diseases, genital infections and abortion. Pathogens' activation of toll-like receptors (TLRs) induces the expression of proinflammatory cytokines and antimicrobial peptides such as cathelicidins. Cathelicidins are presumed to act as endogenous ligands of TLRs, stimulating, in turn, their activation. Cattle's innate immune responses mediated by TLRs and cathelicidins remain undefined. This work aimed to study the interaction mechanisms between TLR7 and a bovine cathelicidin (bovine myeloid antimicrobial peptide (BMAP)28 and their influence on the replication of BoAHV-1 and BoAHV-5. It was shown that in vitro infection by BoAHV-1 and BoAHV-5 induces the innate immune responses associated with TLR7 and BMAP28 in primary cultured bovine fetal lung cells. Pretreatment with TLR7 agonist (imiquimod) or an endogenous cathelicidin inductor (sodium butyrate) enhanced the innate immune response to BoAHV mediated by TLR7 signalling, BMAP28, TNFα, IFNβ and IFNγ. It exerted an antiviral effect, decreasing BoAHV replication. Inhibition of endosomal TLRs and downstream signalling molecules MAPK, NF-κβ, IKK-2, and JNK II counteracted these antiviral effects. Therefore, BMAP28 shows novel roles in modulating the immune response induced by TLR7 activation through signalling pathways involving kinases and NF-κβ. Enhancing TLR7 and endogenous cathelicidin expression could be a complementary tool for the prevention and/or control of BoAHV infections.
Collapse
Affiliation(s)
- Mercedes M Burucúa
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación GISAZ, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - Cecilia A Langellotti
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET) Hurlingham, Buenos Aires, Argentina
| | - Silvina Quintana
- Instituto de Investigaciones de Producción, Sanidad y Ambiente (IIPROSAM), FCEyN, UNMDP-CONICET, Mar del Plata, Buenos Aires, Argentina
| | - Anselmo C Odeón
- Facultad de Ciencias Agrarias, UNMdP, Balcarce, Buenos Aires, Argentina
| | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ana Paula Cutrera
- Laboratorio de Ecología Fisiológica y del Comportamiento, Instituto de Investigaciones Marinas y Costeras (IIMyC), UNMDP-CONICET, Mar del Plata, Buenos Aires, Argentina
| | - Sandra E Pérez
- Centro de Investigaciones Veterinarias de Tandil (CIVETAN) - CONICET, Facultad de Ciencias Veterinarias, UNCPBA, Tandil, Buenos Aires, Argentina
| | - Maia S Marin
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
3
|
Daneshi M, Borowicz PP, Hirchert MR, Entzie YL, Syring JG, King LE, Safain KS, Anas M, Reynolds LP, Ward AK, Dahlen CR, Crouse MS, Caton JS. Influence of maternal nutrition and one-carbon metabolites supplementation on bovine antimicrobial peptides in fetal and maternal tissues. Front Vet Sci 2024; 11:1505427. [PMID: 39720407 PMCID: PMC11666495 DOI: 10.3389/fvets.2024.1505427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024] Open
Abstract
Introduction Maternal nutrition during pregnancy critically influences offspring development and immune function. One-carbon metabolites (OCM) are epigenetic modifiers that may modulate antimicrobial peptide (AMP) expression, which is vital for innate immunity. This study investigated the effects of maternal nutrient restriction and OCM supplementation on mRNA expression of AMP in fetal and maternal lung, mammary gland, and small intestine of beef cattle. Methods Twenty-nine crossbred Angus beef heifers were synchronized for estrus and artificially inseminated. They were assigned to one of four treatments in a 2 × 2 factorial design: nutritional plane [control (CON) vs. restricted (RES)] and OCM supplementation [without OCM (-OCM) or with OCM (+OCM)]. Heifers on the CON diet were fed to gain 0.45 kg/day, while RES heifers were fed to lose 0.23 kg/day. Treatments were applied from day 0 to 63 of gestation, after which all heifers were fed a common diet to gain 0.45 kg/day until day 161 of gestation, when samples were collected. Quantitative RT-qPCR was used to assess mRNA expression of AMP. Results Nutritional plane had no effect (p ≥ 0.24) on mRNA expression of AMP in either the fetus or dams. However, the mRNA expression of cathelicidin5 (CATHL5; p = 0.07) and bovine neutrophil β-defensin5 (BNBD5; p = 0.07) in the fetal lung and mammary gland, respectively, was lower in the +OCM groups compared to the -OCM groups. In the maternal small intestine, the expression of enteric β-defensin (EBD) was lower (p = 0.01) in the +OCM groups compared to the -OCM groups. Additionally, in the maternal lung, there was a tendency (p = 0.06) for an interaction in CATHL5 mRNA expression, with the RES + OCM group showing greater expression compared to the CON + OCM (p = 0.07) and RES - OCM (p = 0.08) groups. Discussion Our findings suggest that while restricted maternal nutrition did not affect mRNA expression of AMP, OCM supplementation modulated AMP expression in both fetal and maternal tissues. Further research is needed to elucidate the mechanisms underlying OCM's impact on AMP expression.
Collapse
Affiliation(s)
- Mojtaba Daneshi
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Pawel P. Borowicz
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Mara R. Hirchert
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Yssi L. Entzie
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Jessica G. Syring
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Layla E. King
- Department of Agriculture and Natural Resources, University of Minnesota Crookston, Crookston, MN, United States
| | - Kazi Sarjana Safain
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Muhammad Anas
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Lawrence P. Reynolds
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Alison K. Ward
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK, Canada
| | - Carl R. Dahlen
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| | - Matthew S. Crouse
- United States Department of Agriculture, Agriculture Research Service, U.S. Meat Animal Research Center, Clay Center, NE, United States
| | - Joel S. Caton
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
4
|
Martínez SR, Caverzan M, Ibarra LE, Aiassa V, Bohl L, Porporatto C, Gómez ML, Chesta CA, Palacios RE. Light-activated conjugated polymer nanoparticles to defeat pathogens associated with bovine mastitis. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 257:112971. [PMID: 38955081 DOI: 10.1016/j.jphotobiol.2024.112971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Bovine mastitis (BM) represents a significant challenge in the dairy industry. Limitations of conventional treatments have prompted the exploration of alternative approaches, such as photodynamic inactivation (PDI). In this study, we developed a PDI protocol to eliminate BM-associated pathogens using porphyrin-doped conjugated polymer nanoparticles (CPN). The PDI-CPN protocol was evaluated in four mastitis isolates of Staphylococcus and in a hyper-biofilm-forming reference strain. The results in planktonic cultures demonstrated that PDI-CPN exhibited a bactericidal profile upon relatively low light doses (∼9.6 J/cm2). Furthermore, following a seven-hour incubation period, no evidence of cellular reactivation was observed, indicating a highly efficient post-photodynamic inactivation effect. The successful elimination of bacterial suspensions encouraged us to test the PDI-CPN protocol on mature biofilms. Treatment using moderate light dose (∼64.8 J/cm2) reduced biofilm biomass and metabolic activity by up to 74% and 88%, respectively. The impact of PDI-CPN therapy on biofilms was investigated using scanning electron microscopy (SEM), which revealed nearly complete removal of the extracellular matrix and cocci. Moreover, ex vivo studies conducted on bovine udder skin demonstrated the efficacy of the therapy in eliminating bacteria from these scaffolds and its potential as a prophylactic method. Notably, the histological analysis of skin revealed no signs of cellular degeneration, suggesting that the protocol is safe and effective for BM treatment. Overall, this study demonstrates the potential of PDI-CPN in treating and preventing BM pathogens. It also provides insights into the effects of PDI-CPN on bacterial growth, metabolism, and survival over extended periods, aiding the development of effective control strategies and the optimization of future treatments.
Collapse
Affiliation(s)
- Sol R Martínez
- Departamento de Química, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Río Cuarto X5804BYA, Córdoba, Argentina.
| | - Matías Caverzan
- Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Río Cuarto X5804BYA, Córdoba, Argentina
| | - Luis E Ibarra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto X5804BYA, Córdoba, Argentina
| | - Virginia Aiassa
- UNITEFA-CONICET, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Luciana Bohl
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB-CONICET), Universidad Nacional de Villa María, Villa María, Argentina. Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María, Villa María, Argentina
| | - Carina Porporatto
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB-CONICET), Universidad Nacional de Villa María, Villa María, Argentina. Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María, Villa María, Argentina
| | - María L Gómez
- Departamento de Química, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Río Cuarto X5804BYA, Córdoba, Argentina
| | - Carlos A Chesta
- Departamento de Química, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Río Cuarto X5804BYA, Córdoba, Argentina
| | - Rodrigo E Palacios
- Departamento de Química, Universidad Nacional de Río Cuarto, Río Cuarto X5804BYA, Córdoba, Argentina; Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Río Cuarto X5804BYA, Córdoba, Argentina.
| |
Collapse
|
5
|
Daneshi M, Caton JS, Caixeta LS, Eftekhari Z, Ward AK. Expression, Regulation, and Function of β-Defensins in the Bovine Mammary Glands: Current Knowledge and Future Perspectives. Animals (Basel) 2023; 13:3372. [PMID: 37958127 PMCID: PMC10650070 DOI: 10.3390/ani13213372] [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: 09/22/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
β-Defensins are cationic antimicrobial peptides (AMPs) that play an important role in the innate immune defense of bovines. They are constitutively expressed in mammary glands and induced differently in response to pathogens. Their expression is influenced by various factors, including hormones, plant-derived compounds, and dietary energy imbalance. The toll-like receptors (TLRs)/nuclear factor-kappa B (NF-κB) pathway plays a crucial role in β-defensin induction, while alternative pathways such as mitogen-activated protein kinase (MAPK) and epigenetic regulation also make substantial contributions. β-Defensins exhibit bactericidal activity against a wide range of pathogens, including two major mastitis pathogens, Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus), primarily through membrane disruption. β-Defensins have low cytotoxicity to host cells and demonstrate immunomodulatory properties, and pathogens also display minimal resistance to these AMPs. Given the increasing concern in antimicrobial resistance, the potential of β-defensins as natural antimicrobials has garnered considerable attention. This article provides an overview of the characteristics of bovine β-defensins, their expression pathways, their mode of action, and factors influencing their expression in the mammary glands of cattle. Additionally, it identifies the current gaps in research within this field and suggests areas that require further investigation. Understanding the regulation and function of β-defensins offers valuable insights to develop effective strategies for strengthening the immune system of mammary glands, reducing the reliance on synthetic antimicrobials, and explore novel natural antimicrobial alternatives.
Collapse
Affiliation(s)
- Mojtaba Daneshi
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Joel S. Caton
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58108, USA
| | - Luciano S. Caixeta
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Zohre Eftekhari
- Biotechnology Department, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Alison K. Ward
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada;
| |
Collapse
|
6
|
Duan H, Wang L, Huangfu M, Li H. The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother 2023; 165:115276. [PMID: 37542852 DOI: 10.1016/j.biopha.2023.115276] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) derived from the fermentation of carbohydrates by gut microbiota play a crucial role in regulating host physiology. Among them, acetate, propionate, and butyrate are key players in various biological processes. Recent research has revealed their significant functions in immune and inflammatory responses. For instance, butyrate reduces the development of interferon-gamma (IFN-γ) generating cells while promoting the development of regulatory T (Treg) cells. Propionate inhibits the initiation of a Th2 immune response by dendritic cells (DCs). Notably, SCFAs have an inhibitory impact on the polarization of M2 macrophages, emphasizing their immunomodulatory properties and potential for therapeutics. In animal models of asthma, both butyrate and propionate suppress the M2 polarization pathway, thus reducing allergic airway inflammation. Moreover, dysbiosis of gut microbiota leading to altered SCFA production has been implicated in prostate cancer progression. SCFAs trigger autophagy in cancer cells and promote M2 polarization in macrophages, accelerating tumor advancement. Manipulating microbiota- producing SCFAs holds promise for cancer treatment. Additionally, SCFAs enhance the expression of hypoxia-inducible factor 1 (HIF-1) by blocking histone deacetylase, resulting in increased production of antibacterial effectors and improved macrophage-mediated elimination of microorganisms. This highlights the antimicrobial potential of SCFAs and their role in host defense mechanisms. This comprehensive review provides an in-depth analysis of the latest research on the functional aspects and underlying mechanisms of SCFAs in relation to macrophage activities in a wide range of diseases, including infectious diseases and cancers. By elucidating the intricate interplay between SCFAs and macrophage functions, this review aims to contribute to the understanding of their therapeutic potential and pave the way for future interventions targeting SCFAs in disease management.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Mingmei Huangfu
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - Hanyang Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
7
|
Tsugami Y, Nii T, Isobe N. Valine Treatment Enhances Antimicrobial Component Production in Mammary Epithelial Cells and the Milk of Lactating Goats Without Influencing the Tight Junction Barrier. J Mammary Gland Biol Neoplasia 2023; 28:3. [PMID: 36801983 PMCID: PMC9938821 DOI: 10.1007/s10911-023-09529-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/24/2023] [Indexed: 02/21/2023] Open
Abstract
The production of antimicrobial components and the formation of less-permeable tight junctions (TJs) are important in the defense system of lactating mammary glands and for safe dairy production. Valine is a branched-chain amino acid that is actively consumed in the mammary glands and promotes the production of major milk components like β-casein; additionally, branched-chain amino acids stimulate antimicrobial component production in the intestines. Therefore, we hypothesized that valine strengthens the mammary gland defense system without influencing milk production. We investigated the effects of valine in vitro using cultured mammary epithelial cells (MECs) and in vivo using the mammary glands of lactating Tokara goats. Valine treatment at 4 mM increased the secretion of S100A7 and lactoferrin as well as the intracellular concentration of β-defensin 1 and cathelicidin 7 in cultured MECs. In addition, an intravenous injection of valine increased S100A7 levels in the milk of Tokara goats without influencing milk yield and milk components (i.e., fat, protein, lactose, and solids). In contrast, valine treatment did not affect TJ barrier function either in vitro or in vivo. These findings indicate that valine enhances antimicrobial component production without influencing milk production and TJ barrier function in lactating mammary glands; thus, valine contributes to safe dairy production.
Collapse
Affiliation(s)
- Yusaku Tsugami
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan.
| | - Takahiro Nii
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan
| | - Naoki Isobe
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan
| |
Collapse
|
8
|
Akhtar M, Naqvi SUAS, Liu Q, Pan H, Ma Z, Kong N, Chen Y, Shi D, Kulyar MFEA, Khan JA, Liu H. Short Chain Fatty Acids (SCFAs) Are the Potential Immunomodulatory Metabolites in Controlling Staphylococcus aureus-Mediated Mastitis. Nutrients 2022; 14:3687. [PMID: 36145063 PMCID: PMC9503071 DOI: 10.3390/nu14183687] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Mastitis is an emerging health concern in animals. An increased incidence of mastitis in dairy cows has been reported in the last few years across the world. It is estimated that up to 20% of cows are suffering from mastitis, causing incompetency in the mucosal immunity and resulting in excessive global economic losses in the dairy industry. Staphylococcus aureus (S. aureus) has been reported as the most common bacterial pathogen of mastitis at clinical and sub-clinical levels. Antibiotics, including penicillin, macrolides, lincomycin, cephalosporins, tetracyclines, chloramphenicol, and methicillin, were used to cure S. aureus-induced mastitis. However, S. aureus is resistant to most antibiotics, and methicillin-resistant S. aureus (MRSA) especially has emerged as a critical health concern. MRSA impairs immune homeostasis leaving the host more susceptible to other infections. Thus, exploring an alternative to antibiotics has become an immediate requirement of the current decade. Short chain fatty acids (SCFAs) are the potent bioactive metabolites produced by host gut microbiota through fermentation and play a crucial role in host/pathogen interaction and could be applied as a potential therapeutic agent against mastitis. The purpose of this review is to summarize the potential mechanism by which SCFAs alleviate mastitis, providing the theoretical reference for the usage of SCFAs in preventing or curing mastitis.
Collapse
Affiliation(s)
- Muhammad Akhtar
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | | | - Qiyao Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong Pan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Ziyu Ma
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Na Kong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Deshi Shi
- Department of Preventive Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Fakhar-e-Alam Kulyar
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jawaria Ali Khan
- Department of Veterinary Medicine, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Huazhen Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
9
|
Li J, Wen Q, Gu F, An L, Yu T. Non-antibiotic strategies for prevention and treatment of internalized Staphylococcus aureus. Front Microbiol 2022; 13:974984. [PMID: 36118198 PMCID: PMC9471010 DOI: 10.3389/fmicb.2022.974984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 12/01/2022] Open
Abstract
Staphylococcus aureus (S. aureus) infections are often difficult to cure completely. One of the main reasons for this difficulty is that S. aureus can be internalized into cells after infecting tissue. Because conventional antibiotics and immune cells have difficulty entering cells, the bacteria can survive long enough to cause recurrent infections, which poses a serious burden in healthcare settings because repeated infections drastically increase treatment costs. Therefore, preventing and treating S. aureus internalization is becoming a research hotspot. S. aureus internalization can essentially be divided into three phases: (1) S. aureus binds to the extracellular matrix (ECM), (2) fibronectin (Fn) receptors mediate S. aureus internalization into cells, and (3) intracellular S. aureus and persistence into cells. Different phases require different treatments. Many studies have reported on different treatments at different phases of bacterial infection. In the first and second phases, the latest research results show that the cell wall-anchored protein vaccine and some microbial agents can inhibit the adhesion of S. aureus to host cells. In the third phase, nanoparticles, photochemical internalization (PCI), cell-penetrating peptides (CPPs), antimicrobial peptides (AMPs), and bacteriophage therapy can effectively eliminate bacteria from cells. In this paper, the recent progress in the infection process and the prevention and treatment of S. aureus internalization is summarized by reviewing a large number of studies.
Collapse
Affiliation(s)
- Jiangbi Li
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Qiangqiang Wen
- Department of Orthopedics, The Affiliated Hospital of Northwest University, Xi’an, China
| | - Feng Gu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Lijuan An
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tiecheng Yu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Tiecheng Yu,
| |
Collapse
|
10
|
Tsugami Y, Suzuki N, Nii T, Isobe N. Sodium Acetate and Sodium Butyrate Differentially Upregulate Antimicrobial Component Production in Mammary Glands of Lactating Goats. J Mammary Gland Biol Neoplasia 2022; 27:133-144. [PMID: 35678903 DOI: 10.1007/s10911-022-09519-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022] Open
Abstract
Short-chain fatty acids activate antimicrobial component production in the intestine. However, their effects on mammary glands remain unclear. We investigated the effects of acetate and butyrate on antimicrobial component production in mammary epithelial cells (MECs) or leukocytes cultured in vitro and in mammary glands of lactating Tokara goats in vivo. Our results showed that butyrate enhanced the production of β-defensin-1 and S100A7 in MECs. Additionally, the infusion of butyrate into mammary glands through the teats enhanced β-defensin-1 and S100A7 concentrations in milk. The infusion of acetate also increased β-defensin-1 and S100A7 concentrations along with those of cathelicidin-2 and interleukin-8, which are produced by leukocytes. Furthermore, acetate promoted cathelicidin-2 and interleukin-8 secretion in leukocytes in vitro. These findings suggest that acetate and butyrate differentially upregulate antimicrobial component production in mammary glands, which could help to develop appropriate treatment for mastitis, thereby reducing economic losses and improving animal welfare in farming environments.
Collapse
Affiliation(s)
- Yusaku Tsugami
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan
| | - Naoki Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan
| | - Takahiro Nii
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan
| | - Naoki Isobe
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama Higashi-Hiroshima, 739-8528, Hiroshima, Japan.
| |
Collapse
|
11
|
Ganesan R, Jeong JJ, Kim DJ, Suk KT. Recent Trends of Microbiota-Based Microbial Metabolites Metabolism in Liver Disease. Front Med (Lausanne) 2022; 9:841281. [PMID: 35615096 PMCID: PMC9125096 DOI: 10.3389/fmed.2022.841281] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome and microbial metabolomic influences on liver diseases and their diagnosis, prognosis, and treatment are still controversial. Research studies have provocatively claimed that the gut microbiome, metabolomics understanding, and microbial metabolite screening are key approaches to understanding liver cancer and liver diseases. An advance of logical innovations in metabolomics profiling, the metabolome inclusion, challenges, and the reproducibility of the investigations at every stage are devoted to this domain to link the common molecules across multiple liver diseases, such as fatty liver, hepatitis, and cirrhosis. These molecules are not immediately recognizable because of the huge underlying and synthetic variety present inside the liver cellular metabolome. This review focuses on microenvironmental metabolic stimuli in the gut-liver axis. Microbial small-molecule profiling (i.e., semiquantitative monitoring, metabolic discrimination, target profiling, and untargeted profiling) in biological fluids has been incompletely addressed. Here, we have reviewed the differential expression of the metabolome of short-chain fatty acids (SCFAs), tryptophan, one-carbon metabolism and bile acid, and the gut microbiota effects are summarized and discussed. We further present proof-of-evidence for gut microbiota-based metabolomics that manipulates the host's gut or liver microbes, mechanosensitive metabolite reactions and potential metabolic pathways. We conclude with a forward-looking perspective on future attention to the "dark matter" of the gut microbiota and microbial metabolomics.
Collapse
|
12
|
Zhang M, Wang Y, Zhao X, Liu C, Wang B, Zhou J. Mechanistic basis and preliminary practice of butyric acid and butyrate sodium to mitigate gut inflammatory diseases: a comprehensive review. Nutr Res 2021; 95:1-18. [PMID: 34757305 DOI: 10.1016/j.nutres.2021.08.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 01/02/2023]
Abstract
A key event featured in the early stage of chronic gut inflammatory diseases is the disordered recruitment and excess accumulation of immune cells in the gut lamina propria. This process is followed by the over-secretion of pro-inflammatory factors and the prolonged overactive inflammatory responses. Growing evidence has suggested that gut inflammatory diseases may be mitigated by butyric acid (BA) or butyrate sodium (NaB). Laboratory studies show that BA and NaB can enhance gut innate immune function through G-protein-mediated signaling pathways while mitigating the overactive inflammatory responses by inhibiting histone deacetylase. The regulatory effects may occur in both epithelial enterocytes and the immune cells in the lamina propria. Prior to further clinical trials, comprehensive literature reviews and rigid examination concerning the underlying mechanism are necessary. To this end, we collected and reviewed 197 published reports regarding the mechanisms, bioactivities, and clinical effects of BA and NaB to modulate gut inflammatory diseases. Our review found insufficient evidence to guarantee the safety of clinical practice of BA and NaB, either by anal enema or oral administration of capsule or tablet. The safety of clinical use of BA and NaB should be further evaluated. Alternatively, dietary patterns rich in "fruits, vegetables and beans" may be an effective and safe approach to prevent gut inflammatory disease, which elevates gut microbiota-dependent production of BA. Our review provides a comprehensive reference to future clinical trials of BA and NaB to treat gut inflammatory diseases.
Collapse
Affiliation(s)
- Mingbao Zhang
- Department of Gastroenterology and Hepatology, Second Hospital of Shandong University, Shandong University, 250012 China
| | - Yanan Wang
- Department of Gastroenterology and Hepatology, Second Hospital of Shandong University, Shandong University, 250012 China
| | - Xianqi Zhao
- School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China
| | - Chang Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China
| | - Baozhen Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China.
| | - Jun Zhou
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China.
| |
Collapse
|
13
|
Gut microbiome and metabolic response in non-alcoholic fatty liver disease. Clin Chim Acta 2021; 523:304-314. [PMID: 34666025 DOI: 10.1016/j.cca.2021.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/19/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
Fatty liver disease (FLD) is one of the largest burdens to human health worldwide and is associated with gut microbiome and metabolite stability. Engineered liver tissues have shown promise in restoring liver functions in non-alcoholic FLD (NAFLD), hepatitis and cirrhosis. Fatty liver, largely noted in obesity and hepatic cancer, is highly fatal and has led to a global increase in death rates. It is associated with complex metabolic reprogramming too. A standard approach to therapy in the newly diagnosed setting includes surgery or identification of biomarkers/ metabolites for therapeutic purposes, which ultimately focus on improvement of liver health in patients. As such there are no standard procedures for patient care, but depending on the severity, systemic therapy with either genomic, proteomic or metabolomic profiling form potential options. Better comparisons and study of underlying mechanisms in gut microbiome-based metabolic functions in obesity are urgently required. Today, an emerging field, focusing on metabolomic approaches and metabolic phenotyping, involved in high-throughput identification of metabolome in obesity and gut disorders, is involved in biomarker and metabolite identification. There are supporting technologies and approaches in NAFLD that throw light on the metabolites and gut microbiome, and also on the understanding of the risk factors of obesity along with liver cancer metabolic reaction networks. We discuss the current state of NAFLD metabolites, gut micro-environmental changes, and the further challenges in digital metabolomics profiling. Innovative clinical trial designs, with biomarker-enrichment strategies that are required to improve the outcome of NAFLD in patients are also discussed.
Collapse
|
14
|
Machado MG, Sencio V, Trottein F. Short-Chain Fatty Acids as a Potential Treatment for Infections: a Closer Look at the Lungs. Infect Immun 2021; 89:e0018821. [PMID: 34097474 PMCID: PMC8370681 DOI: 10.1128/iai.00188-21] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.
Collapse
Affiliation(s)
- Marina Gomes Machado
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
15
|
Ochoa-Zarzosa A, Báez-Magaña M, Guzmán-Rodríguez JJ, Flores-Alvarez LJ, Lara-Márquez M, Zavala-Guerrero B, Salgado-Garciglia R, López-Gómez R, López-Meza JE. Bioactive Molecules From Native Mexican Avocado Fruit (Persea americana var. drymifolia): A Review. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2021; 76:133-142. [PMID: 33704631 DOI: 10.1007/s11130-021-00887-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/04/2021] [Indexed: 06/12/2023]
Abstract
Avocado (Persea americana Mill.) is a tree native from central and eastern México that belongs to the Lauraceae family. Avocado has three botanical varieties known as Mexican (P. americana var. drymifolia), West Indian (P. americana var. americana), and Guatemalan (P. americana var. guatemalensis). It is an oil-rich fruit appreciated worldwide because of its nutritional value and the content of bioactive molecules. Several avocado molecules show attractive activities of interest in medicine. Avocado fatty acids have beneficial effects on cardiovascular disease risk factors. Besides, this fruit possesses a high content of carotenoids and phenolic compounds with possible antifungal, anti-cancer and antioxidant activities. Moreover, several metabolites have been reported with anti-inflammatory effects. Also, an unsaponifiable fraction of avocado in combination with soybean oil is used for the treatment of osteoarthritis. The Mexican variety is native from México and is characterized by the anise aroma in leaves and by small thin-skinned fruits of rich flavor and excellent quality. However, the study of the bioactive molecules of the fruit has not been addressed in detail. In this work, we achieved a literature review on the inflammatory, immunomodulatory and cytotoxic properties of long-chain fatty acids and derivatives from Mexican avocado seed. Also, the antioxidant and anti-inflammatory properties of the oil extracted from the avocado seed are referred. Finally, the antimicrobial, immunomodulatory, and cytotoxic activities of some antimicrobial peptides expressed in the fruit are reviewed.
Collapse
Affiliation(s)
- 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 Carr, Morelia-Zinapécuaro, Posta Veterinaria, Michoacán, C.P. 58893, Morelia, México
| | - Marisol Báez-Magaña
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carr, Morelia-Zinapécuaro, Posta Veterinaria, Michoacán, C.P. 58893, Morelia, México
| | - Jaquelina Julia Guzmán-Rodríguez
- Campus Irapuato-Salamanca, División de Ciencias de la Vida, Posgrado en Biociencias, Universidad de Guanajuato, 36500, Irapuato, Guanajuato, México
| | - Luis José Flores-Alvarez
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carr, Morelia-Zinapécuaro, Posta Veterinaria, Michoacán, C.P. 58893, Morelia, México
| | - Mónica Lara-Márquez
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carr, Morelia-Zinapécuaro, Posta Veterinaria, Michoacán, C.P. 58893, Morelia, México
| | - Baruc Zavala-Guerrero
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58004, Morelia, Michoacán, México
| | - Rafael Salgado-Garciglia
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58004, Morelia, Michoacán, México
| | - Rodolfo López-Gómez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, C.P. 58004, Morelia, Michoacán, México
| | - Joel Edmundo López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carr, Morelia-Zinapécuaro, Posta Veterinaria, Michoacán, C.P. 58893, Morelia, México.
| |
Collapse
|
16
|
Hu X, Mu R, Xu M, Yuan X, Jiang P, Guo J, Cao Y, Zhang N, Fu Y. Gut microbiota mediate the protective effects on endometritis induced by Staphylococcus aureus in mice. Food Funct 2021; 11:3695-3705. [PMID: 32307472 DOI: 10.1039/c9fo02963j] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Endometritis, the inflammation of the endometrial lining caused by bacterial pathogens, is associated with reproductive failure. Recent studies have shown that gut microbiota play an important role in infectious diseases. However, the roles of the gut microbiota in endometritis remain unclear. Here, we assessed the effects and mechanisms of the gut microbiota during endometritis induced by Staphylococcus aureus (S. aureus). A mouse gut microbiota-dysbiosis model was established by a mixture of antibiotics (Abx) and subsequently, a model of endometritis was established by the uterine perfusion of S. aureus. Fecal microbiota transplantation (FMT) was performed to evaluate the relationship between gut microbiota and endometritis. The results showed that the mice with gut microbiota-dysbiosis developed uterine inflammation, while this inflammatory response of the uterus was alleviated in mice with FMT to gut microbiota-dysbiosis. In addition, S. aureus-induced endometritis was greater in severity in the mice with gut dysbiosis as compared to the untreated mice. Moreover, these effects were reversed in mice with FMT to the gut microbiota-dysbiosis. GC-MS analysis demonstrated that the levels of short-chain fatty acids (SCFAs) in the feces of mice with gut microbiota-dysbiosis significantly decreased and pretreatment with sodium butyrate or sodium propionate increased the concentrations of butyrate or propionate in both the circulation and uterine tissues, thereby reducing the severity of endometritis induced by S. aureus. In addition, the increased pathogen load in the uteri of the mice with gut microbiota-dysbiosis was associated with a reduction in the phagocytic ability and responsiveness of neutrophils. In conclusion, the gut microbiota offer a protective effect against S. aureus-induced endometritis by regulating the levels of SCFAs and maintaining the phagocytic ability and responsiveness of neutrophils.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | | | - Mingyue Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Xin Yuan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Peng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Jian Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People Republic of China.
| |
Collapse
|
17
|
Orellano MS, Bohl LP, Breser ML, Isaac P, Falcone RD, Porporatto C. A comparative study of antimicrobial activity of differently-synthesized chitosan nanoparticles against bovine mastitis pathogens. SOFT MATTER 2021; 17:694-703. [PMID: 33216104 DOI: 10.1039/d0sm01179g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The greatest concern in dairy farming nowadays is bovine mastitis (BM), which results mainly from bacterial colonization of the mammary gland. Antibiotics are the most widely used strategy for its prevention and treatment, but overuse has led to growing antimicrobial resistance. Pathogens have also developed other mechanisms to persist in the udder, such as biofilm formation and internalization into bovine epithelial cells. New therapies are therefore needed to reduce or replace antibiotic therapies. In a previous study, we found that chitosan nanoparticles (Ch-NPs) have considerable potential for the treatment of BM. The aim of the present study was to evaluate the antimicrobial activity of differently-synthesized Ch-NPs against BM pathogens and their toxicity in bovine cells in vitro, to further explore the attributes of Ch-NPs for the prevention and treatment of intramammary infections. We also looked into their ability to inhibit biofilm formation and prevent the internalization of S. aureus into mammary epithelial cells. Finally, since an interesting approach for BM prevention is to enhance the host's immune response, we studied whether Ch-NPs could promote the release of pro-inflammatory cytokines in mammary epithelial cells. The results reveal that the bactericidal effect of Ch-NPs on BM pathogens and their ability to inhibit biofilm formation are size-dependent, with smaller particles being more efficient. In contrast, their effect on the viability of the cell lines is not size-dependent and all samples tested were non-toxic. The smallest Ch-NPs successfully prevented the internalization of S. aureus into the cells, but did not promote the production of pro-inflammatory cytokines. These findings make it possible to conclude that Ch-NPs are a great bactericidal agent which can prevent the main mechanisms developed by BM pathogens to persist in the udder.
Collapse
Affiliation(s)
- M Soledad Orellano
- Centro de Investigaciones y Transferencia de Villa María (CIT-VM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Villa María (UNVM), Villa María, Argentina. and Instituto para el Desarrollo Agroindustrial y de la Salud (IDAS), CONICET. Departamento de Química, Universidad Nacional de Río Cuarto (UNRC), Agencia Postal # 3. C.P. X5804BYA, Río Cuarto, Argentina
| | - Luciana P Bohl
- Centro de Investigaciones y Transferencia de Villa María (CIT-VM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Villa María (UNVM), Villa María, Argentina.
| | - María L Breser
- Centro de Investigaciones y Transferencia de Villa María (CIT-VM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Villa María (UNVM), Villa María, Argentina.
| | - Paula Isaac
- Centro de Investigaciones y Transferencia de Villa María (CIT-VM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Villa María (UNVM), Villa María, Argentina.
| | - R Darío Falcone
- Instituto para el Desarrollo Agroindustrial y de la Salud (IDAS), CONICET. Departamento de Química, Universidad Nacional de Río Cuarto (UNRC), Agencia Postal # 3. C.P. X5804BYA, Río Cuarto, Argentina
| | - Carina Porporatto
- Centro de Investigaciones y Transferencia de Villa María (CIT-VM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Villa María (UNVM), Villa María, Argentina.
| |
Collapse
|
18
|
Li H, Yu Q, Li T, Shao L, Su M, Zhou H, Qu J. Rumen Microbiome and Metabolome of Tibetan Sheep ( Ovis aries) Reflect Animal Age and Nutritional Requirement. Front Vet Sci 2020; 7:609. [PMID: 32984417 PMCID: PMC7492597 DOI: 10.3389/fvets.2020.00609] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/28/2020] [Indexed: 01/26/2023] Open
Abstract
The rumen microbiota plays an important role in animal functional attributes. These microbes are indispensable for the normal physiological development of the rumen, and may also convert the plant polysaccharides from grass into available milk and meat, making it highly valuable to humans. Exploring the microbial composition and metabolites of rumen across developmental stages is important for understanding ruminant nutrition and metabolism. However, relatively few reports have investigated the microbiome and metabolites across developmental stages in ruminants. Using 16S rRNA gene sequnecing, metabolomics and high-performance liquid chromatography techniques, we compared the rumen microbiota, metabolites and short chain fatty acids (SCFAs) between lambs and sub-adult Tibetan sheep (Ovis aries) from Qinghai-Tibetan Plateau. Bacteroidetes and Spirochaetae were enriched in sub-adult sheep, while Firmicutes and Tenericutes were more abundant in young individuals. The sub-adult individuals had higher alpha diversity values than those in young sheep. Metabolomics analysis showed that the content of essential amino acids and related gene functional pathways in rumen were different between the lambs and sub-adult population. L-Leucine that participates in valine, leucine and isoleucine biosynthesis was more abundant in the lambs, while phenylethylamine that takes part in phenylalanine metabolism was more enriched in the sub-adults. Both rumen microbial community structures and metabolite profiles were impacted by age, but rumen SCFA concentration was relatively stable between different age stages. Some specific microbes (e.g., Clostridium and Ruminococcaceae) were positively associated with L-Leucine but negatively correlated with phenylethylamine, implying that rumen microbes may play different roles for metabolite production at different ages. Mantel test analysis showed that rumen microbiota was significantly correlated with metabolomics and SCFA profiles. Our results indicates the close relationship between microbial composition and metabolites, and also reveal different nutritional requirement for different ages in ruminants, thus having important significance for regulating animal nutrition and metabolism by microbiome intervention.
Collapse
Affiliation(s)
- Huan Li
- School of Public Health, Lanzhou University, Lanzhou, China.,Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China
| | - Qiaoling Yu
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Tongtong Li
- Department of Applied Biology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liye Shao
- Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, China
| | - Ming Su
- Central South Inventory and Planning Institute of National Forestry and Grassland Administration, Changsha, China
| | - Huakun Zhou
- Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China.,Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Jiapeng Qu
- Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China.,Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| |
Collapse
|
19
|
Chang PV. Chemical Mechanisms of Colonization Resistance by the Gut Microbial Metabolome. ACS Chem Biol 2020; 15:1119-1126. [PMID: 31895538 DOI: 10.1021/acschembio.9b00813] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The gut microbiome, the collection of 100 trillion microorganisms that resides in the intestinal lumen, plays major roles in modulating host physiology. One well-established function of the gut microbiota is that of colonization resistance or the ability of the microbial collective to protect the host against enteric pathogens. Although evidence suggests that these microbes may outcompete some pathogens, there remains a lack of mechanistic understanding that underlies this competitive exclusion. In recent years, there has been great interest in small-molecule metabolites that are produced by the gut microbiota and in understanding how these molecules regulate host-pathogen interactions. In this review, we briefly summarize these findings by focusing on several classes of metabolites that mediate this important process. Understanding these host-microbe interactions in the gut may lead to identification of potential candidates for the development of prophylactics and therapeutics for many infectious diseases that are impacted by the gut microbiome.
Collapse
Affiliation(s)
- Pamela V. Chang
- Department of Microbiology and Immunology, Cornell Institute of Host-Microbe Interactions & Disease, and Cornell Center for Immunology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
20
|
Zhang K, Hussain T, Wang J, Li M, Wang W, Ma X, Liao Y, Yao J, Song Y, Liang Z, Zhou X, Xu L. Sodium Butyrate Abrogates the Growth and Pathogenesis of Mycobacterium bovis via Regulation of Cathelicidin (LL37) Expression and NF-κB Signaling. Front Microbiol 2020; 11:433. [PMID: 32265874 PMCID: PMC7096352 DOI: 10.3389/fmicb.2020.00433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium bovis is the causative agent of bovine tuberculosis, has been identified a serious threat to human population. It has been found that sodium butyrate (NaB), the inhibitor of histone deacetylase, can promote the expression of cathelicidin (LL37) and help the body to resist a variety of injuries. In the current study, we investigate the therapeutic effect of NaB on the regulation of host defense mechanism against M. bovis infection. We found an increased expression of LL37 in M. bovis infected THP-1 cells after NaB treatment. In contrast, NaB treatment significantly down-regulated the expression of Class I HDAC in THP-1 cells infected with M. bovis. Additionally, NaB reduced the expression of phosphorylated P65 (p-P65) and p-IκBα, indicating the inhibition of nuclear factor-κB (NF-κB) signaling. Furthermore, we found that NaB treatment reduced the production of inflammatory cytokines (IL-1β, TNF-α, and IL-10) and a key anti-apoptotic marker protein Bcl-2 in THP-1 cell infected with M. bovis. Notably, mice showed high resistance to M. bovis infection after NaB treatment. The reduction of viable M. bovis bacilli indicates that NaB-induced inhibition of M. bovis infection mediated by upregulation of LL37 and inhibition of NF-κB signaling pathway. These observations illustrate that NaB mediate protective immune responses against M. bovis infection. Overall, these results suggest that NaB can be exploited as a therapeutic strategy for the control of M. bovis in animals and human beings.
Collapse
Affiliation(s)
- Kai Zhang
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Tariq Hussain
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.,College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Jie Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Mengying Li
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Wenjia Wang
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Xiaojing Ma
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Yi Liao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiao Yao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yinjuan Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhengmin Liang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lihua Xu
- School of Agriculture, Ningxia University, Yinchuan, China
| |
Collapse
|
21
|
Mu D, Luan Y, Wang L, Gao Z, Yang P, Jing S, Wang Y, Xiang H, Wang T, Wang D. The combination of salvianolic acid A with latamoxef completely protects mice against lethal pneumonia caused by methicillin-resistant Staphylococcus aureus. Emerg Microbes Infect 2020; 9:169-179. [PMID: 31969071 PMCID: PMC7006784 DOI: 10.1080/22221751.2020.1711817] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Staphylococcus aureus (S. aureus), especially methicillin-resistant Staphylococcus aureus (MRSA), is a major cause of pneumonia, resulting in severe morbidity and mortality in adults and children. Sortase A (SrtA), which mediates the anchoring of cell surface proteins in the cell wall, is an important virulence factor of S. aureus. Here, we found that salvianolic acid A (Sal A), which is a natural product that does not affect the growth of S. aureus, could inhibit SrtA activity (IC50 = 5.75 μg/ml) and repress the adhesion of bacteria to fibrinogen, the anchoring of protein A to cell wall, the biofilm formation, and the ability of S. aureus to invade A549 cells. Furthermore, in vivo studies demonstrated that Sal A treatment reduced inflammation and protected mice against lethal pneumonia caused by MRSA. More significantly, full protection (a survival rate of 100%) was achieved when Sal A was administered in combination with latamoxef. Together, these results indicate that Sal A could be developed into a promising therapeutic drug to combat MRSA infections while limiting resistance development.
Collapse
Affiliation(s)
- Dan Mu
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Yongxin Luan
- Department of Neurosurgery, First Hospital of Jilin University, Jilin University, Changchun, People's Republic of China
| | - Lin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Zeyuan Gao
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Panpan Yang
- Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun, People's Republic of China
| | - Shisong Jing
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Yanling Wang
- College of Animal Science, Jilin University, Changchun, People's Republic of China.,Qingdao Vland biological Limited co., LTD, Qingdao, People's Republic of China
| | - Hua Xiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, People's Republic of China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| | - Dacheng Wang
- College of Animal Science, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
22
|
Raimondi I, Izzo L, Tunesi M, Comar M, Albani D, Giordano C. Organ-On-A-Chip in vitro Models of the Brain and the Blood-Brain Barrier and Their Value to Study the Microbiota-Gut-Brain Axis in Neurodegeneration. Front Bioeng Biotechnol 2020; 7:435. [PMID: 31998702 PMCID: PMC6965718 DOI: 10.3389/fbioe.2019.00435] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
We are accumulating evidence that intestinal microflora, collectively named gut microbiota, can alter brain pathophysiology, but researchers have just begun to discover the mechanisms of this bidirectional connection (often referred to as microbiota-gut-brain axis, MGBA). The most noticeable hypothesis for a pathological action of gut microbiota on the brain is based on microbial release of soluble neurotransmitters, hormones, immune molecules and neuroactive metabolites, but this complex scenario requires reliable and controllable tools for its causal demonstration. Thanks to three-dimensional (3D) cultures and microfluidics, engineered in vitro models could improve the scientific knowledge in this field, also from a therapeutic perspective. This review briefly retraces the main discoveries linking the activity of gut microbiota to prevalent brain neurodegenerative disorders, and then provides a deep insight into the state-of-the-art for in vitro modeling of the brain and the blood-brain barrier (BBB), two key players of the MGBA. Several brain and BBB microfluidic devices have already been developed to implement organ-on-a-chip solutions, but some limitations still exist. Future developments of organ-on-a-chip tools to model the MGBA will require an interdisciplinary approach and the synergy with cutting-edge technologies (for instance, bioprinting) to achieve multi-organ platforms and support basic research, also for the development of new therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilaria Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Manola Comar
- SSD of Advanced Translational Microbiology, IRCCS “Burlo Garofolo”, Department of Medical Sciences (DMS), University of Trieste, Trieste, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| |
Collapse
|
23
|
Chen J, Zhai Z, Long H, Yang G, Deng B, Deng J. Inducible expression of defensins and cathelicidins by nutrients and associated regulatory mechanisms. Peptides 2020; 123:170177. [PMID: 31704211 DOI: 10.1016/j.peptides.2019.170177] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Host defense peptides (HDPs) are crucial components of the body's first line of defense that protect organisms from infections and mediate immune responses. Defensins and cathelicidins are the two most important families of HDPs in mammals. In this review, we summarize the nutrients that are involved in inducible expression of endogenous defensins and cathelicidins. In addition, the mitogen-activated protein kinases (MAPK), nuclear factor kappa B (NF-κB) and histone deacetylase (HDAC) signaling pathways that play vital roles in the induction of defensin and cathelicidin expression are highlighted. Endogenous defensins and cathelicidins induced by nutrients may be potential alternatives to antibiotic treatments against infection and diseases. This review mainly focuses on the inducible expression and regulatory mechanisms of defensins and cathelicidins in multiple species by different nutrients and the potential applications of defensin- and cathelicidin-inducing nutrients.
Collapse
Affiliation(s)
- Jialuo Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenya Zhai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hongrong Long
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guangming Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
24
|
Lipid-Rich Extract from Mexican Avocado Seed ( Persea americana var. drymifolia) Reduces Staphylococcus aureus Internalization and Regulates Innate Immune Response in Bovine Mammary Epithelial Cells. J Immunol Res 2019; 2019:7083491. [PMID: 31612151 PMCID: PMC6757280 DOI: 10.1155/2019/7083491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Bovine mammary epithelial cells (bMECs) are capable of initiating an innate immune response (IIR) to invading bacteria. Staphylococcus aureus is not classically an intracellular pathogen, although it has been shown to be internalized into bMECs. S. aureus internalizes into nonprofessional phagocytes, which allows the evasion of the IIR and turns antimicrobial therapy unsuccessful. An alternative treatment to control this pathogen is the modulation of the innate immune response of the host. The Mexican avocado (Persea americana var. drymifolia) is a source of molecules with anti-inflammatory and immunomodulatory properties. Hence, we analyze the effect of a lipid-rich extract from avocado seed (LEAS) on S. aureus internalization into bMECs and their innate immunity response. The effects of LEAS (1-500 ng/ml) on the S. aureus growth and bMEC viability were assessed by turbidimetry and MTT assays, respectively. LEAS did not show neither antimicrobial nor cytotoxic effects. S. aureus internalization into bMECs was analyzed by gentamicin protection assays. Interestingly, LEAS (1-200 ng/ml) decreased bacterial internalization (60-80%) into bMECs. This effect correlated with NO production and the induction of the gene expression of IL-10, while the expression of the proinflammatory cytokine TNF-α was reduced. These effects could be related to the inhibition of MAPK p38 (∼60%) activation by LEAS. In conclusion, our results showed that LEAS inhibits the S. aureus internalization into bMECs and modulates the IIR, which indicates that avocado is a source of metabolites for control of mastitis pathogens.
Collapse
|
25
|
Targeting gut microbiota as a possible therapy for mastitis. Eur J Clin Microbiol Infect Dis 2019; 38:1409-1423. [PMID: 31079312 DOI: 10.1007/s10096-019-03549-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023]
Abstract
Mastitis, a disease that affects both dairy herds and humans, is recognized as the most common source of losses in the dairy industry. Antibiotics have been used for years as the primary treatment for mastitis. However, abuse of antibiotics has led to the emergence of resistant strains and the presence of drug residues and has increased the difficulty of curing this disease. In addition, antibiotics kill most of the microbes that are present in the digestive tract, leading to imbalances in the gut microbiome and destruction of the ecosystem that is normally present in the gut. Gut microbiota play an important role in the host's health and could be considered the "second brain" of the body. In recent years, the gut microbiota and their metabolites, including lipopolysaccharide (LPS) and short-chain fatty acids (SCFAs), have been shown to participate in the development of mastitis. LPS is the main component of the cell walls of gram-negative bacteria. Overproduction of rumen-derived LPS injures the rumen epithelium, resulting in the entry of LPS into the blood and damaged liver function; once in the blood, it circulates into the mammary gland, increasing blood-barrier permeability and leading to mammary gland inflammation. SCFAs, which are produced by gut microbiota as fermentation products, have a protective effect on mammary gland inflammatory responses and help maintain the function of the blood-milk barrier. Recently, increasing attention has been focused on the use of probiotics as a promising alternative for the treatment of mastitis. This review summarizes the effects of the gut microbiome and its metabolites on mastitis as well as the current of probiotics in mastitis. This work may provide a valuable theoretical foundation for the development of fresh ideas for the prevention and treatment of mastitis.
Collapse
|
26
|
Park JW, Kim HY, Kim MG, Jeong S, Yun CH, Han SH. Short-chain Fatty Acids Inhibit Staphylococcal Lipoprotein-induced Nitric Oxide Production in Murine Macrophages. Immune Netw 2019; 19:e9. [PMID: 31089436 PMCID: PMC6494764 DOI: 10.4110/in.2019.19.e9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/02/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus, a Gram-positive pathogen, can cause severe inflammation in humans, leading to various life-threatening diseases. The lipoprotein is a major virulence factor in S. aureus-induced infectious diseases and is responsible for excessive inflammatory mediators such as nitric oxide (NO). Short-chain fatty acids (SCFAs) including butyrate, propionate, and acetate are microbial metabolites in the gut that are known to have anti-inflammatory effects in the host. In this study, we investigated the effects of SCFAs on S. aureus lipoprotein (Sa.LPP)-induced NO production in mouse macrophages. Butyrate and propionate, but not acetate, inhibited Sa.LPP-induced production of NO in RAW 264.7 cells and bone marrow-derived macrophages. Butyrate and propionate inhibited Sa.LPP-induced expression of inducible NO synthase (iNOS). However, acetate did not show such effects under the same conditions. Furthermore, butyrate and propionate, but not acetate, inhibited Sa.LPP-induced activation of NF-κB, expression of IFN-β, and phosphorylation of STAT1, which are essential for inducing transcription of iNOS in macrophages. In addition, butyrate and propionate induced histone acetylation at lysine residues in the presence of Sa.LPP in RAW 264.7 cells. Moreover, Sa.LPP-induced NO production was decreased by histone deacetylase (HDAC) inhibitors. Collectively, these results suggest that butyrate and propionate ameliorate the inflammatory responses caused by S. aureus through the inhibition of NF-κB, IFN-β/STAT1, and HDAC, resulting in attenuated NO production in macrophages.
Collapse
Affiliation(s)
- Jeong Woo Park
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Korea
| | - Hyun Young Kim
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Korea
| | - Min Geun Kim
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Korea
| | - Soyoung Jeong
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
27
|
Martínez-Cortés I, Acevedo-Domínguez NA, Olguin-Alor R, Cortés-Hernández A, Álvarez-Jiménez V, Campillo-Navarro M, Sumano-López HS, Gutiérrez-Olvera L, Martínez-Gómez D, Maravillas-Montero JL, Loor JJ, García-Zepeda EA, Soldevila G. Tilmicosin modulates the innate immune response and preserves casein production in bovine mammary alveolar cells during Staphylococcus aureus infection. J Anim Sci 2019; 97:644-656. [PMID: 30517644 DOI: 10.1093/jas/sky463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/01/2018] [Indexed: 12/15/2022] Open
Abstract
Tilmicosin is an antimicrobial agent used to treat intramammary infections against Staphylococcus aureus and has clinical anti-inflammatory effects. However, the mechanism by which it modulates the inflammatory process in the mammary gland is unknown. We evaluated the effect of tilmicosin treatment on the modulation of the mammary innate immune response after S. aureus infection and its effect on casein production in mammary epithelial cells. To achieve this goal, we used immortalized mammary epithelial cells (MAC-T), pretreated for 12 h or treated with tilmicosin after infection with S. aureus (ATCC 27543). Our data showed that tilmicosin decreases intracellular infection (P < 0.01) and had a protective effect on MAC-T reducing apoptosis after infection by 80% (P < 0.01). Furthermore, tilmicosin reduced reactive oxygen species (ROS) (P < 0.01), IL-1β (P < 0.01), IL-6 (P < 0.01), and TNF-α (P < 0.05) production. In an attempt to investigate the signaling pathways involved in the immunomodulatory effect of tilmicosin, mitogen-activated protein kinase (MAPK) phosphorylation was measured by fluorescent-activated cell sorting. Pretreatment with tilmicosin increased ERK1/2 (P < 0.05) but decreased P38 phosphorylation (P < 0.01). In addition, the anti-inflammatory effect of tilmicosin helped to preserve casein synthesis in mammary epithelial cells (P < 0.01). This result indicates that tilmicosin could be an effective modulator inflammation in the mammary gland. Through regulation of MAPK phosphorylation, ROS production and pro-inflammatory cytokine secretion tilmicosin can provide protection from cellular damage due to S. aureus infection and help to maintain normal physiological functions of the bovine mammary epithelial cell.
Collapse
Affiliation(s)
- Ismael Martínez-Cortés
- Chemokine Biology Research Laboratory, UNAM, Ciudad de México, Mexico.,Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Naray A Acevedo-Domínguez
- Chemokine Biology Research Laboratory, UNAM, Ciudad de México, Mexico.,Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Roxana Olguin-Alor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Arimelek Cortés-Hernández
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Violeta Álvarez-Jiménez
- Chemokine Biology Research Laboratory, UNAM, Ciudad de México, Mexico.,Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Marcia Campillo-Navarro
- Laboratorio de Inmunología Integrativa-INER, Ismael Cosio Villegas. Ciudad de México, Mexico
| | | | | | | | | | - Juan J Loor
- Mammalian NutriPhysioGenomics-University of Illinois, Urbana, IL
| | - Eduardo A García-Zepeda
- Chemokine Biology Research Laboratory, UNAM, Ciudad de México, Mexico.,Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, Ciudad de México, Mexico
| |
Collapse
|
28
|
Zhou Z, Yang H, Li H, Li X, Li X, Wu B, Tian S, Wu J, Wang Z, Hu S. Sodium butyrate ameliorates Corynebacterium pseudotuberculosis infection in RAW264.7 macrophages and C57BL/6 mice. Microb Pathog 2019; 131:144-149. [PMID: 30965088 DOI: 10.1016/j.micpath.2019.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/23/2019] [Accepted: 04/05/2019] [Indexed: 12/22/2022]
Abstract
Corynebacterium pseudotuberculosis (CP) infection in livestock has become highly difficult to control. To decrease the incidence of CP infection, the supplementation of feed with non-antibiotic antibacterial substances is a potential approach. The aim of this study was to assess the effects of sodium butyrate (NaB), a potential alternative to antibiotics, on CP infection in RAW264.7 macrophages and C57BL/6 mice. Our data showed that NaB (2 mM) significantly ameliorated CPinfection in RAW264.7 macrophages and decreased the bacterial load in the spleens of infected mice. By real-time PCR, we found that NaB induced significant decreases in zinc-dependent superoxide dismutase (sodC) and tip protein C (spaC) expression in CP from infected-RAW264.7 cells and in phospholipase D (pld) and spaC expression in CP from the spleens of infected mice. NaB treatment significantly up-regulated cathelicidin-related antimicrobial peptide (cramp) expression in spleens of mice infected with CP. Furthermore, NaB alleviated histopathological changes in spleens of CP-infected mice. In conclusion, NaB ameliorated CP infection in RAW264.7 macrophages and C57BL/6 mice, and these effects may be related to the modulation of sodC, spaC, pld, and cramp expression.
Collapse
Affiliation(s)
- Zuoyong Zhou
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China; Veterinary Science Engineering Research Center of Chongqing, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Haoyue Yang
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Hexian Li
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Xiaoxia Li
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Xiao Li
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Bi Wu
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Shangquan Tian
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Junjun Wu
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Zhiying Wang
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China; Veterinary Science Engineering Research Center of Chongqing, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| | - Shijun Hu
- College of Animal Science, Rongchang Campus of Southwest University, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China; Veterinary Science Engineering Research Center of Chongqing, No. 160 Xueyuan Road, Rongchang District, Chongqing, 402460, China.
| |
Collapse
|
29
|
Frutis-Murillo M, Sandoval-Carrillo MA, Alva-Murillo N, Ochoa-Zarzosa A, López-Meza JE. Immunomodulatory molecules regulate adhesin gene expression in Staphylococcus aureus: Effect on bacterial internalization into bovine mammary epithelial cells. Microb Pathog 2019; 131:15-21. [PMID: 30930221 DOI: 10.1016/j.micpath.2019.03.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/11/2019] [Accepted: 03/25/2019] [Indexed: 11/26/2022]
Abstract
Staphylococcus aureus is a major pathogen of subclinical bovine mastitis that usually is chronic and recurrent, which has been related to its ability to internalize into bovine mammary epithelial cells (bMECs). Previously, we reported that short and medium fatty acids and cholecalciferol reduce S. aureus internalization into pretreated-bMECs with these molecules suggesting a role as immunomodulatory agents. Hence, we assessed the role of sodium butyrate (NaB), sodium octanoate (NaO) and cholecalciferol on S. aureus adhesin expression and its internalization into bMECs. S. aureus pre-treated 2 h with 0.5 mM or 2 mM NaB showed a reduction in internalization into bMECs (∼35% and ∼55%; respectively), which coincided with a down-regulated expression of clumping factor B (ClfB). Also, the S. aureus internalization reduction by 2 mM NaB (2 h) agreed with a down-regulated expression of sdrC. Moreover, the 2 mM NaB (24 h) pre-treatment induced bacterial internalization (∼3-fold), which was related with an up-regulation of spa, clfB and sdrC genes. Also, NaO (0.25 mM and 1 mM) only reduced S. aureus internalization when bacteria were grown 2 h with this molecule but there was no relationship with adhesin expression. In addition, cholecalciferol (50 nM) reduced bacteria internalization at similar levels (∼50%) when bacteria were grown 2 and 24 h in broth supplemented with this compound, which correlated with spa and sdrC mRNA expression down-regulated at 2 h, and fnba and clfB mRNA expression decreased at 24 h. In conclusion, our data support the fact that fatty acids and cholecalciferol regulate adhesin gene expression as well as bacteria internalization in nonprofessional phagocytic cells, which may lead to development of anti-virulence agents for control of pathogens.
Collapse
Affiliation(s)
- Minerva Frutis-Murillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Marcelo Alejandro Sandoval-Carrillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Nayeli Alva-Murillo
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Mexico
| | - Alejandra Ochoa-Zarzosa
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, 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, Morelia, Michoacán, Mexico.
| |
Collapse
|
30
|
Silva LG, Ferguson BS, Avila AS, Faciola AP. Sodium propionate and sodium butyrate effects on histone deacetylase (HDAC) activity, histone acetylation, and inflammatory gene expression in bovine mammary epithelial cells. J Anim Sci 2019; 96:5244-5252. [PMID: 30252114 DOI: 10.1093/jas/sky373] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase (HDAC) inhibition attenuates inflammation in rodents and short-chain fatty acids (SCFAs) are effective HDAC inhibitors. Therefore, the objective of this study was to evaluate the role of the SCFAs sodium propionate (SP) and sodium butyrate (SB) as HDAC-dependent regulators of inflammatory gene expression in bovine mammary epithelial cells (MAC-Ts). We postulated that SP and SB would decrease inflammation in MAC-Ts by inhibiting HDAC activity and increasing histone H3 acetylation and consequently decreasing inflammatory gene expression. For this study, MAC-Ts stimulated with lipopolysaccharide (LPS) were used as a model for bovine mammary epithelial cell inflammation. MAC-Ts were cultured in a basal medium. Cell lysates were incubated with SP or SB (0 to 5 mM) for 2 h prior to HDAC substrates incubation for an additional 2 h and HDACs activity was determined. Next, cells were pretreated with SP or SB (0 to 3.0 mM) for 2 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and assessed for histone H3 acetylation. Then, cells were pretreated with SP or SB (1 mM) for 24 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and RNA was isolated for inflammatory gene expression evaluation by PCR array and gene validation was performed using quantitative real-time PCR. One-way ANOVA followed by Tukey post hoc analysis was conducted and statistical significance set at P < 0.05. SP and SB concentration-dependently and selectively inhibited class I HDAC activity, which differed between SCFAs, where SB inhibited (P < 0.05) HDACs 2, 3, and 8, while SP inhibited (P < 0.05) HDACs 2 and 8. Histone H3 acetylation was concentration-dependently increased by SCFAs and likewise the differential regulation of HDAC activity, SCFAs effected differently histone H3 acetylation, where SB increased (P < 0.05) H3K9/14, H3K18 and H3K27 acetylation, while SP increased (P < 0.05) H3K9/14 and H3K18 acetylation. However, SCFAs did not decrease (P > 0.05) overall inflammatory gene expression. Under our experimental conditions, findings suggest that in MAC-Ts, SCFAs regulate epigenetic markers on nucleosomal DNA in addition to regulation of inflammatory gene events independent of HDAC activity. Nevertheless, examination of SCFAs and/or HDACs inhibitors in bovine mammary gland is worth being further investigated to delineate the potential impact of HDAC inhibition and histones hyperacetylation on mammary gland tissue inflammation.
Collapse
Affiliation(s)
- Lorrayny G Silva
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Bradley S Ferguson
- Department Agriculture, Nutrition, and Veterinary Sciences, University of Nevada, Reno, NV
| | - Andre S Avila
- Department of Animal Sciences, University of Florida, Gainesville, FL.,Agricultural Science Center, Universidade Estadual do Oeste do Parana, Marechal Candido Rondon, PR, Brazil
| | - Antonio P Faciola
- Department of Animal Sciences, University of Florida, Gainesville, FL
| |
Collapse
|
31
|
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
|
32
|
Development of a Cell-Based High-Throughput Screening Assay to Identify Porcine Host Defense Peptide-Inducing Compounds. J Immunol Res 2018; 2018:5492941. [PMID: 30581875 PMCID: PMC6276403 DOI: 10.1155/2018/5492941] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/19/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022] Open
Abstract
Novel alternatives to antibiotics are needed for the swine industry, given increasing restrictions on subtherapeutic use of antibiotics. Augmenting the synthesis of endogenous host defense peptides (HDPs) has emerged as a promising antibiotic-alternative approach to disease control and prevention. To facilitate the identification of HDP inducers for swine use, we developed a stable luciferase reporter cell line, IPEC-J2/PBD3-luc, through permanent integration of a luciferase reporter gene driven by a 1.1 kb porcine β-defensin 3 (PBD3) gene promoter in porcine IPEC-J2 intestinal epithelial cells. Such a stable reporter cell line was employed in a high-throughput screening of 148 epigenetic compounds and 584 natural products, resulting in the identification of 41 unique hits with a minimum strictly standardized mean difference (SSMD) value of 3.0. Among them, 13 compounds were further confirmed to give at least a 5-fold increase in the luciferase activity in the stable reporter cell line, with 12 being histone deacetylase (HDAC) inhibitors. Eight compounds were subsequently observed to be comparable to sodium butyrate in inducing PBD3 mRNA expression in parental IPEC-J2 cells in the low micromolar range. Six HDAC inhibitors including suberoylanilide hydroxamine (SAHA), HC toxin, apicidin, panobinostat, SB939, and LAQ824 were additionally found to be highly effective HDP inducers in a porcine 3D4/31 macrophage cell line. Besides PBD3, other HDP genes such as PBD2 and cathelicidins (PG1–5) were concentration-dependently induced by those compounds in both IPEC-J2 and 3D4/31 cells. Furthermore, the antibacterial activities of 3D4/31 cells were augmented following 24 h exposure to HDAC inhibitors. In conclusion, a cell-based high-throughput screening assay was developed for the discovery of porcine HDP inducers, and newly identified HDP-inducing compounds may have potential to be developed as alternatives to antibiotics for applications in swine and possibly other animal species.
Collapse
|
33
|
Aabdin ZU, Bilal MS, Dai H, Abaker JA, Liu X, Benazir S, Yan J, Shen X. NOD1/NF-κB signaling pathway inhibited by sodium butyrate in the mammary gland of lactating goats during sub-acute ruminal acidosis. Microb Pathog 2018; 122:58-62. [DOI: 10.1016/j.micpath.2018.05.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/09/2018] [Accepted: 05/28/2018] [Indexed: 01/01/2023]
|
34
|
Su YN, Wu P, Feng L, Jiang WD, Jiang J, Zhang YA, Figueiredo-Silva C, Zhou XQ, Liu Y. The improved growth performance and enhanced immune function by DL-methionyl-DL-methionine are associated with NF-κB and TOR signalling in intestine of juvenile grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2018; 74:101-118. [PMID: 29292200 DOI: 10.1016/j.fsi.2017.12.051] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 06/07/2023]
Abstract
The present study investigated the effects of dietary DL-methionyl-DL-methionine (Met-Met) on growth performance, intestinal immune function and the underlying signalling molecules in juvenile grass carp (Ctenopharyngodon idella). Fish were fed one DL-methionine (DL-Met) group (2.50 g/kg diet) and six graded levels of Met-Met groups (0, 0.79, 1.44, 1.84, 2.22 and 2.85 g/kg diet) for 10 weeks, and then challenged with Aeromonas hydrophila for 14 days. Results indicated that the optimal Met-Met supplementation: (1) increased fish growth performance, intestinal lysozyme (LZ) and acid phosphatase (ACP) activities, complement (C3 and C4) and immunoglobulin M (IgM) contents, up-regulated hepcidin, liver expressed antimicrobial peptide 2A (LEAP-2A), LEAP-2B, β-defensin-1 and Mucin2 mRNA levels; (2) down-regulated tumour necrosis factor α (TNF-α), interferon γ2 (IFN-γ2), interleukin 1β (IL-1β), IL-8 [only in the distal intestine (DI)], IL-12p35, IL-12p40 and IL-15 (not IL-17D) mRNA levels partially related to the down-regulation of IκB kinase β (IKKβ) and IKKγ (rather than IKKα), nuclear factor kappa B (NF-κB) p65 and c-Rel (rather than NF-κB p52) mRNA levels and the up-regulation of inhibitor of κBα (IκBα) mRNA levels; (3) up-regulated IL-4/13A, IL-4/13B, IL-6, IL-10, IL-11 and transforming growth factor (TGF)-β1 (not TGF-β2) mRNA levels partially associated with the target of rapamycin (TOR) signalling pathway [TOR/ribosomal protein S6 kinases 1 (S6K1), eIF4E-binding proteins (4E-BP)] in three intestinal segments of juvenile grass carp. These results suggest that Met-Met supplementation improves growth and intestinal immune function in fish. Furthermore, according to a positive effect, the optimal Met-Met supplementation was superior to the optimal DL-Met supplementation at improving the growth performance and enhancing the intestinal immune function in fish. Finally, based on percent weight gain (PWG), protection against enteritis morbidity and immune index (LZ activity), the optimal Met-Met supplementation for juvenile grass carp was estimated as 1.61, 1.64 and 1.68 g/kg diet, respectively, as the basal diet contains 8.03 g/kg total sulfur amino acids (TSAA) (4.26 g methionine/kg and 3.77 g cysteine/kg).
Collapse
Affiliation(s)
- Yue-Ning Su
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Claudia Figueiredo-Silva
- Evonik Nutrition & Care GmbH, NC, 10-B531, Postfach 1345, Rodenbacher Chausse 4, 63404 Hanau, Germany
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China.
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China.
| |
Collapse
|
35
|
Dietary modulation of endogenous host defense peptide synthesis as an alternative approach to in-feed antibiotics. ACTA ACUST UNITED AC 2018; 4:160-169. [PMID: 30140755 PMCID: PMC6104571 DOI: 10.1016/j.aninu.2018.01.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 01/01/2018] [Accepted: 01/06/2018] [Indexed: 12/11/2022]
Abstract
Traditionally, antibiotics are included in animal feed at subtherapeutic levels for growth promotion and disease prevention. However, recent links between in-feed antibiotics and a rise in antibiotic-resistant pathogens have led to a ban of all antibiotics in livestock production by the European Union in January 2006 and a removal of medically important antibiotics in animal feeds in the United States in January 2017. An urgent need arises for antibiotic alternatives capable of maintaining animal health and productivity without triggering antimicrobial resistance. Host defense peptides (HDP) are a critical component of the animal innate immune system with direct antimicrobial and immunomodulatory activities. While in-feed supplementation of recombinant or synthetic HDP appears to be effective in maintaining animal performance and alleviating clinical symptoms in the context of disease, dietary modulation of the synthesis of endogenous host defense peptides has emerged as a cost-effective, antibiotic-alternative approach to disease control and prevention. Several different classes of small-molecule compounds have been found capable of promoting HDP synthesis. Among the most efficacious compounds are butyrate and vitamin D. Moreover, butyrate and vitamin D synergize with each other in enhancing HDP synthesis. This review will focus on the regulation of HDP synthesis by butyrate and vitamin D in humans, chickens, pigs, and cattle and argue for potential application of HDP-inducing compounds in antibiotic-free livestock production.
Collapse
|
36
|
Wei Z, Xiao C, Guo C, Zhang X, Wang Y, Wang J, Yang Z, Fu Y. Sodium acetate inhibits Staphylococcus aureus internalization into bovine mammary epithelial cells by inhibiting NF-κB activation. Microb Pathog 2017; 107:116-121. [PMID: 28351710 DOI: 10.1016/j.micpath.2017.03.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
Abstract
Bovine mastitis is one of the most costly and prevalent disease affecting dairy cows worldwide. It was reported that Staphylococcus aureus could internalize into bovine mammary epithelial cells (bMEC) and induce mastitis. Some short chain fatty acids (SCFA) have shown to suppress S. aureus invasion into bMEC and regulate antimicrobial peptides expression. But it has not been evaluated that sodium acetate has the similar effect. The aim of this study was to investigate the effect of sodium acetate on the invasion of bovine mammary epithelial cells (bMEC) by S. aureus. Gentamicin protection assay showed that the invasion of S. aureus into bMEC was inhibited by sodium acetate in a dose-dependent manner. Sodium acetate (0.25-5 mM) did not affect S. aureus growth and bMEC viability. The TAP gene level was decreased, while the BNBD5 mRNA level was enhanced in sodium acetate treated bMEC. In sodium acetate treated and S. aureus challenged bMEC, the TAP gene expression was increased and BNBD5 gene expression was not modified at low concentrations, but decreased at high concentrations. The Nitric oxide (NO) production of bMEC after S. aureus stimulation was decreased by sodium acetate treatment. Furthermore, sodium acetate treatment suppressed S. aureus-induced NF-κB activation in bMEC in a dose manner. In conclusion, our results suggested that sodium acetate exerts an inhibitory property on S. aureus internalization and modulates antimicrobial peptides gene expression.
Collapse
Affiliation(s)
- Zhengkai Wei
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Chong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Changming Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Xu Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Yanan Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Jingjing Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Zhengtao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China; Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China.
| |
Collapse
|
37
|
Kao MS, Huang S, Chang WL, Hsieh MF, Huang CJ, Gallo RL, Huang CM. Microbiome precision editing: Using PEG as a selective fermentation initiator against methicillin-resistant Staphylococcus aureus. Biotechnol J 2017; 12. [PMID: 27982519 DOI: 10.1002/biot.201600399] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 12/02/2016] [Accepted: 12/13/2016] [Indexed: 12/20/2022]
Abstract
Recent creation of a Unified Microbiome Initiative (UMI) has the aim of understanding how microbes interact with each other and with us. When pathogenic Staphylococcus aureus infects the skin, the interplay between S. aureus and skin commensal bacteria occurs. Our previous data revealed that skin commensal bacteria can mediate fermentation against the growth of USA300, a community-acquired methicillin-resistant S. aureus MRSA. By using a fermentation process with solid media on a small scale, we define poly(ethylene glycol) dimethacrylate (PEG-DMA) as a selective fermentation initiator which can specifically intensify the probiotic ability of skin commensal Staphylococcus epidermidis bacteria. At least five short-chain fatty acids including acetic, butyric and propionic acids with anti-USA300 activities are produced by PEG-DMA fermentation of S. epidermidis. Furthermore, the S. epidermidis-laden PEG-DMA hydrogels effectively decolonized USA300 in skin wounds in mice. The PEG-DMA and its derivatives may become novel biomaterials to specifically tailor the human skin microbiome against invading pathogens.
Collapse
Affiliation(s)
- Ming-Shan Kao
- Department of Dermatology, University of California, San Diego, California, USA.,Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Stephen Huang
- Surface Bioadvances Inc., San Diego, California, USA
| | - Wei-Lin Chang
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Ming-Fa Hsieh
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chun-Jen Huang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, California, USA
| | - Chun-Ming Huang
- Department of Dermatology, University of California, San Diego, California, USA.,Moores Cancer Center, University of California, San Diego, California, USA
| |
Collapse
|
38
|
Chen H, Chen T, Giudici P, Chen F. Vinegar Functions on Health: Constituents, Sources, and Formation Mechanisms. Compr Rev Food Sci Food Saf 2016; 15:1124-1138. [PMID: 33401833 DOI: 10.1111/1541-4337.12228] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 11/30/2022]
Abstract
Vinegars are one of only a few acidic condiments throughout the world. Vinegars can mainly be considered grain vinegars and fruit vinegars, according to the raw materials used. Both grain vinegars and fruit vinegars, which are fermented by traditional methods, possess a variety of physiological functions, such as antibacteria, anti-infection, antioxidation, blood glucose control, lipid metabolism regulation, weight loss, and anticancer activities. The antibacteria and anti-infection abilities of vinegars are mainly due to the presence of organic acids, polyphenols, and melanoidins. The polyphenols and melanoidins also provide the antioxidant abilities of vinegars, which are produced from the raw materials and fermentation processes, respectively. The blood glucose control, lipid metabolism regulation, and weight loss capabilities from vinegars are mainly due to acetic acid. Besides caffeoylsophorose (inhibits disaccharidase) and ligustrazine (improves blood circulation), other functional ingredients present in vinegars provide certain health benefits as well. Regarding anticancer activities, several grain vinegars strongly inhibit the growth of some cancer cells in vivo or in vitro, but related functional ingredients remain largely unknown, except tryptophol in Japanese black soybean vinegar. Considering the discovering of various functional ingredients and clarifying their mechanisms, some vinegars could be functional foods or even medicines, depending on a number of proofs that demonstrate these constituents can cure chronic diseases such as diabetes or cardiovascular problems.
Collapse
Affiliation(s)
- Hengye Chen
- Key Laboratory of Environment Correlative Dietology and College of Food Science and Technology, Huazhong Agricultural Uni, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Tao Chen
- Key Laboratory of Environment Correlative Dietology and College of Food Science and Technology, Huazhong Agricultural Uni, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Paolo Giudici
- Dept. of Life Sciences, Uni. of Modena and Reggio Emilia, Via Amendola, 2, 42122, Reggio Emilia, Italy
| | - Fusheng Chen
- Key Laboratory of Environment Correlative Dietology and College of Food Science and Technology, Huazhong Agricultural Uni, Wuhan, 430070, Hubei Province, People's Republic of China
| |
Collapse
|
39
|
Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology 2016; 5:e73. [PMID: 27195116 PMCID: PMC4855267 DOI: 10.1038/cti.2016.17] [Citation(s) in RCA: 867] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are bacterial fermentation products, which are chemically composed by a carboxylic acid moiety and a small hydrocarbon chain. Among them, acetic, propionic and butyric acids are the most studied, presenting, respectively, two, three and four carbons in their chemical structure. These metabolites are found in high concentrations in the intestinal tract, from where they are uptaken by intestinal epithelial cells (IECs). The SCFAs are partially used as a source of ATP by these cells. In addition, these molecules act as a link between the microbiota and the immune system by modulating different aspects of IECs and leukocytes development, survival and function through activation of G protein coupled receptors (FFAR2, FFAR3, GPR109a and Olfr78) and by modulation of the activity of enzymes and transcription factors including the histone acetyltransferase and deacetylase and the hypoxia-inducible factor. Considering that, it is not a surprise, the fact that these molecules and/or their targets are suggested to have an important role in the maintenance of intestinal homeostasis and that changes in components of this system are associated with pathological conditions including inflammatory bowel disease, obesity and others. The aim of this review is to present a clear and updated description of the effects of the SCFAs derived from bacteria on host immune system, as well as the molecular mechanisms involved on them.
Collapse
|
40
|
Díaz-Murillo V, Medina-Estrada I, López-Meza JE, Ochoa-Zarzosa A. Defensin γ-thionin from Capsicum chinense has immunomodulatory effects on bovine mammary epithelial cells during Staphylococcus aureus internalization. Peptides 2016; 78:109-18. [PMID: 26939717 DOI: 10.1016/j.peptides.2016.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/20/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022]
Abstract
β-Defensins are members of the antimicrobial peptide superfamily that are produced in various species from different kingdoms, including plants. Plant defensins exhibit primarily antifungal activities, unlike those from animals that exhibit a broad-spectrum antimicrobial action. Recently, immunomodulatory roles of mammal β-defensins have been observed to regulate inflammation and activate the immune system. Similar roles for plant β-defensins remain unknown. In addition, the regulation of the immune system by mammalian β-defensins has been studied in humans and mice models, particularly in immune cells, but few studies have investigated these peptides in epithelial cells, which are in intimate contact with pathogens. The aim of this work was to evaluate the effect of the chemically synthesized β-defensin γ-thionin from Capsicum chinense on the innate immune response of bovine mammary epithelial cells (bMECs) infected with Staphylococcus aureus, the primary pathogen responsible for bovine mastitis, which is capable of living within bMECs. Our results indicate that γ-thionin at 0.1 μg/ml was able to reduce the internalization of S. aureus into bMECs (∼50%), and it also modulates the innate immune response of these cells by inducing the mRNA expression (∼5-fold) and membrane abundance (∼3-fold) of Toll-like receptor 2 (TLR2), as well as by inducing genes coding for the pro-inflammatory cytokines TNF-α and IL-1β (∼14 and 8-fold, respectively) before and after the bacterial infection. γ-Thionin also induces the expression of the mRNA of anti-inflammatory cytokine IL-10 (∼12-fold). Interestingly, the reduction in bacterial internalization coincides with the production of other antimicrobial products by bMECs, such as NO before infection, and the secretion into the medium of the endogenous antimicrobial peptide DEFB1 after infection. The results from this work support the potential use of β-defensins from plants as immunomodulators of the mammalian innate immune response.
Collapse
Affiliation(s)
- Violeta Díaz-Murillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carr. Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico, Mexico
| | - 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 Carr. Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico, 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 Carr. Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico, 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 Carr. Morelia-Zinapécuaro, Posta Veterinaria, C.P. 58893, Morelia, Michoacán, Mexico, Mexico.
| |
Collapse
|
41
|
Robinson K, Deng Z, Hou Y, Zhang G. Regulation of the Intestinal Barrier Function by Host Defense Peptides. Front Vet Sci 2015; 2:57. [PMID: 26664984 PMCID: PMC4672242 DOI: 10.3389/fvets.2015.00057] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/02/2015] [Indexed: 12/17/2022] Open
Abstract
Intestinal barrier function is achieved primarily through regulating the synthesis of mucins and tight junction (TJ) proteins, which are critical for maintaining optimal gut health and animal performance. An aberrant expression of TJ proteins results in increased paracellular permeability, leading to intestinal and systemic disorders. As an essential component of innate immunity, host defense peptides (HDPs) play a critical role in mucosal defense. Besides broad-spectrum antimicrobial activities, HDPs promotes inflammation resolution, endotoxin neutralization, wound healing, and the development of adaptive immune response. Accumulating evidence has also indicated an emerging role of HDPs in barrier function and intestinal homeostasis. HDP deficiency in the intestinal tract is associated with barrier dysfunction and dysbiosis. Several HDPs were recently shown to enhance mucosal barrier function by directly inducing the expression of multiple mucins and TJ proteins. Consistently, dietary supplementation of HDPs often leads to an improvement in intestinal morphology, production performance, and feed efficiency in livestock animals. This review summarizes current advances on the regulation of epithelial integrity and homeostasis by HDPs. Major signaling pathways mediating HDP-induced mucin and TJ protein synthesis are also discussed. As an alternative strategy to antibiotics, supplementation of exogenous HDPs or modulation of endogenous HDP synthesis may have potential to improve intestinal barrier function and animal health and productivity.
Collapse
Affiliation(s)
- Kelsy Robinson
- Department of Animal Science, Oklahoma State University , Stillwater, OK , USA
| | - Zhuo Deng
- Department of Animal Science, Oklahoma State University , Stillwater, OK , USA
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University , Wuhan , China
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University , Stillwater, OK , USA ; Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University , Wuhan , China
| |
Collapse
|
42
|
The activation of the TLR2/p38 pathway by sodium butyrate in bovine mammary epithelial cells is involved in the reduction of Staphylococcus aureus internalization. Mol Immunol 2015; 68:445-55. [PMID: 26471700 DOI: 10.1016/j.molimm.2015.09.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 09/26/2015] [Accepted: 09/29/2015] [Indexed: 01/31/2023]
Abstract
Staphylococcus aureus is an etiological agent of human and animal diseases, and it is able to internalize into non-professional phagocytic cells (i.e. bovine mammary epithelial cells, bMECs), which is an event that is related to chronic and recurrent infections. bMECs contribute to host innate immune responses (IIR) through TLR pathogen recognition, whereby TLR2 is the most relevant for S. aureus. In a previous report, we showed that sodium butyrate (NaB, 0.5mM), which is a short chain fatty acid (SCFA), reduced S. aureus internalization into bMECs by modulating their IIR. However, the molecular mechanism of this process has not been described, which was the aim of this study. The results showed that the TLR2 membrane abundance (MA) and mRNA expression were induced by 0.5mM NaB ∼1.6-fold and ∼1.7-fold, respectively. Additionally, 0.5mM NaB induced p38 phosphorylation, but not JNK1/2 or ERK1/2 phosphorylation in bMECs, which reached the baseline when the bMECs were S. aureus-challenged. Additionally, bMECs that were treated with 0.5mM NaB (24h) showed activation of 8 transcriptional factors (AP-1, E2F-1, FAST-1, MEF-1, EGR, PPAR, ER and CBF), which were partially reverted when the bMECs were S. aureus-challenged. Additionally, 0.5mM NaB (24h) up-regulated mRNA expression of the antimicrobial peptides, TAP (∼4.8-fold), BNBD5 (∼3.2-fold) and BNBD10 (∼2.6-fold). Notably, NaB-treated and S. aureus-challenged bMECs increased the mRNA expression of all of the antimicrobial peptides that were evaluated, and this was evident for LAP and BNBD5. In the NaB-treated bMECs, we did not detect significant expression changes for IL-1β and IL-6 and only TNF-α, IL-10 and IL-8 were induced. Interestingly, the NaB-treated and S. aureus-challenged bMECs maintained the anti-inflammatory response that was induced by this SCFA. In conclusion, our results suggest that 0.5mM NaB activates bMECs via TLR2/p38, which leads to improved antimicrobial defense before/after pathogen invasion, and NaB may exert anti-inflammatory effects during infection.
Collapse
|
43
|
Wei Z, Fu Y, Zhou E, Tian Y, Yao M, Li Y, Yang Z, Cao Y. Effects of niacin on Staphylococcus aureus internalization into bovine mammary epithelial cells by modulating NF-κB activation. Microb Pathog 2014; 71-72:62-67. [DOI: 10.1016/j.micpath.2014.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/15/2014] [Accepted: 03/20/2014] [Indexed: 11/16/2022]
|
44
|
|
45
|
Thymol inhibits Staphylococcus aureus internalization into bovine mammary epithelial cells by inhibiting NF-κB activation. Microb Pathog 2014; 71-72:15-9. [PMID: 24583152 DOI: 10.1016/j.micpath.2014.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 11/23/2022]
Abstract
Bovine mastitis is one of the most costly and prevalent diseases in the dairy industry and is characterised by inflammatory and infectious processes. Staphylococcus aureus (S. aureus), a Gram-positive organism, is a frequent cause of subclinical, chronic mastitis. Thymol, a monocyclic monoterpene compound isolated from Thymus vulgaris, has been reported to have antibacterial properties. However, the effect of thymol on S. aureus internalization into bovine mammary epithelial cells (bMEC) has not been investigated. In this study, we evaluated the effect of thymol on S. aureus internalization into bMEC, the expression of tracheal antimicrobial peptide (TAP) and β-defensin (BNBD5), and the inhibition of NF-κB activation in bMEC infected with S. aureus. Our results showed that thymol (16-64 μg/ml) could reduce the internalization of S. aureus into bMEC and down-regulate the mRNA expression of TAP and BNBD5 in bMEC infected with S. aureus. In addition, thymol was found to inhibit S. aureus-induced nitric oxide (NO) production in bMEC and suppress S. aureus-induced NF-κB activation in a dose-dependent manner. In conclusion, these results indicated that thymol inhibits S. aureus internalization into bMEC by inhibiting NF-κB activation.
Collapse
|
46
|
Elangovan S, Pathania R, Ramachandran S, Ananth S, Padia RN, Lan L, Singh N, Martin PM, Hawthorn L, Prasad PD, Ganapathy V, Thangaraju M. The niacin/butyrate receptor GPR109A suppresses mammary tumorigenesis by inhibiting cell survival. Cancer Res 2013; 74:1166-78. [PMID: 24371223 DOI: 10.1158/0008-5472.can-13-1451] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
GPR109A, a G-protein-coupled receptor, is activated by niacin and butyrate. Upon activation in colonocytes, GPR109A potentiates anti-inflammatory pathways, induces apoptosis, and protects against inflammation-induced colon cancer. In contrast, GPR109A activation in keratinocytes induces flushing by activation of Cox-2-dependent inflammatory signaling, and the receptor expression is upregulated in human epidermoid carcinoma. Thus, depending on the cellular context and tissue, GPR109A functions either as a tumor suppressor or a tumor promoter. However, the expression status and the functional implications of this receptor in the mammary epithelium are not known. Here, we show that GPR109A is expressed in normal mammary tissue and, irrespective of the hormone receptor status, its expression is silenced in human primary breast tumor tissues, breast cancer cell lines, and in tumor tissues of three different murine mammary tumor models. Functional expression of this receptor in human breast cancer cell lines decreases cyclic AMP production, induces apoptosis, and blocks colony formation and mammary tumor growth. Transcriptome analysis revealed that GPR109A activation inhibits genes, which are involved in cell survival and antiapoptotic signaling, in human breast cancer cells. In addition, deletion of Gpr109a in mice increased tumor incidence and triggered early onset of mammary tumorigenesis with increased lung metastasis in MMTV-Neu mouse model of spontaneous breast cancer. These findings suggest that GPR109A is a tumor suppressor in mammary gland and that pharmacologic induction of this gene in tumor tissues followed by its activation with agonists could be an effective therapeutic strategy to treat breast cancer.
Collapse
Affiliation(s)
- Selvakumar Elangovan
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Biostatistics and Epidemiology, and Pathology; Cancer Center; Vision Science Discovery Institute, Georgia Regents University; Augusta, Georgia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Farrerol regulates antimicrobial peptide expression and reduces Staphylococcus aureus internalization into bovine mammary epithelial cells. Microb Pathog 2013; 65:1-6. [PMID: 24036182 DOI: 10.1016/j.micpath.2013.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 08/07/2013] [Accepted: 08/12/2013] [Indexed: 12/13/2022]
Abstract
Mastitis, defined as inflammation of the mammary gland, is an infectious disease with a major economic influence on dairy industry. Staphylococcus aureus is a common gram-positive pathogen that frequently causes subclinical, chronic infection of the mammary gland in dairy cows. Farrerol, a traditional Chinese medicine isolated from rhododendron, has been shown to have anti-bacterial activity. However, the effect of farrerol on S. aureus infection in mammary epithelium has not been studied in detail. The aim of this study was to investigate the effect of farrerol on the invasion of bovine mammary epithelial cells (bMEC) by S. aureus. The expression of antimicrobial peptide genes by bMEC were assessed in the presence or absence of S. aureus infection. Our results demonstrated that farrerol (4-16 μg/ml) reduced > 55% the internalization of S. aureus into bMEC. We also found that farrerol was able to down-regulate the mRNA expression of tracheal antimicrobial peptide (TAP) and bovine neutrophil β-defensin 5 (BNBD5) in bMEC infected with S. aureus. The Nitric oxide (NO) production of bMEC after S. aureus stimulation was decreased by farrerol treatment. Furthermore, farrerol treatment suppressed S. aureus-induced NF-κB activation in bMEC. These results demonstrated that farrerol modulated TAP and BNBD5 gene expression in mammary gland, enhances bMEC defense against S. aureus infection and could be useful in protection against bovine mastitis.
Collapse
|
48
|
Zeng X, Sunkara LT, Jiang W, Bible M, Carter S, Ma X, Qiao S, Zhang G. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. PLoS One 2013; 8:e72922. [PMID: 24023657 PMCID: PMC3758276 DOI: 10.1371/journal.pone.0072922] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/13/2013] [Indexed: 01/08/2023] Open
Abstract
Dietary modulation of the synthesis of endogenous host defense peptides (HDPs) represents a novel antimicrobial approach for disease control and prevention, particularly against antibiotic-resistant infections. However, HDP regulation by dietary compounds such as butyrate is species-dependent. To examine whether butyrate could induce HDP expression in pigs, we evaluated the expressions of a panel of porcine HDPs in IPEC-J2 intestinal epithelial cells, 3D4/31 macrophages, and primary monocytes in response to sodium butyrate treatment by real-time PCR. We revealed that butyrate is a potent inducer of multiple, but not all, HDP genes. Porcine β-defensin 2 (pBD2), pBD3, epididymis protein 2 splicing variant C (pEP2C), and protegrins were induced markedly in response to butyrate, whereas pBD1 expression remained largely unaltered in any cell type. Additionally, a comparison of the HDP-inducing efficacy among saturated free fatty acids of different aliphatic chain lengths revealed that fatty acids containing 3–8 carbons showed an obvious induction of HDP expression in IPEC-J2 cells, with butyrate being the most potent and long-chain fatty acids having only a marginal effect. We further investigated a panel of butyrate analogs for their efficacy in HDP induction, and found glyceryl tributyrate, benzyl butyrate, and 4-phenylbutyrate to be comparable with butyrate. Identification of butyrate and several analogs with a strong capacity to induce HDP gene expression in pigs provides attractive candidates for further evaluation of their potential as novel alternatives to antibiotics in augmenting innate immunity and disease resistance of pigs.
Collapse
Affiliation(s)
- Xiangfang Zeng
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Lakshmi T. Sunkara
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Weiyu Jiang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Megan Bible
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Scott Carter
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, United States of America
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, United States of America
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
49
|
Lucero CM, Fallert Junecko B, Klamar CR, Sciullo LA, Berendam SJ, Cillo AR, Qin S, Sui Y, Sanghavi S, Murphey-Corb MA, Reinhart TA. Macaque paneth cells express lymphoid chemokine CXCL13 and other antimicrobial peptides not previously described as expressed in intestinal crypts. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1320-8. [PMID: 23803902 PMCID: PMC3754526 DOI: 10.1128/cvi.00651-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 06/12/2013] [Indexed: 12/30/2022]
Abstract
CXCL13 is a constitutively expressed chemokine that controls migration of immune cells to lymphoid follicles. Previously, we found CXCL13 mRNA levels increased in rhesus macaque spleen tissues during AIDS. This led us to examine the levels and locations of CXCL13 by detailed in situ methods in cynomolgus macaque lymphoid and intestinal tissues. Our results revealed that there were distinct localization patterns of CXCL13 mRNA compared to protein in germinal centers. These patterns shifted during the course of simian immunodeficiency virus (SIV) infection, with increased mRNA expression within and around follicles during AIDS compared to uninfected or acutely infected animals. Unexpectedly, CXCL13 expression was also found in abundance in Paneth cells in crypts throughout the small intestine. Therefore, we expanded our analyses to include chemokines and antimicrobial peptides (AMPs) not previously demonstrated to be expressed by Paneth cells in intestinal tissues. We examined the expression patterns of multiple chemokines, including CCL25, as well as α-defensin 6 (DEFA6), β-defensin 2 (BDEF2), rhesus θ-defensin 1 (RTD-1), and Reg3γ in situ in intestinal tissues. Of the 10 chemokines examined, CXCL13 was unique in its expression by Paneth cells. BDEF2, RTD-1, and Reg3γ were also expressed by Paneth cells. BDEF2 and RTD-1 previously have not been shown to be expressed by Paneth cells. These findings expand our understanding of mucosal immunology, innate antimicrobial defenses, homeostatic chemokine function, and host protective mechanisms against microbial translocation.
Collapse
Affiliation(s)
- Carissa M. Lucero
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Beth Fallert Junecko
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cynthia R. Klamar
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lauren A. Sciullo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stella J. Berendam
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony R. Cillo
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shulin Qin
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yongjun Sui
- National Cancer Institute, Bethesda, Maryland, USA
| | - Sonali Sanghavi
- King Edward Memorial Hospital and Research Center, Rasta Peth, Pune, India
| | - Michael A. Murphey-Corb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Todd A. Reinhart
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Effects of sodium octanoate on innate immune response of mammary epithelial cells during Staphylococcus aureus internalization. BIOMED RESEARCH INTERNATIONAL 2012; 2013:927643. [PMID: 23509807 PMCID: PMC3591121 DOI: 10.1155/2013/927643] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/18/2012] [Accepted: 10/26/2012] [Indexed: 11/24/2022]
Abstract
Bovine mammary epithelial cells (bMECs) are capable of initiating an innate immune response to invading bacteria. Short chain fatty acids can reduce Staphylococcus aureus internalization into bMEC, but it has not been evaluated if octanoic acid (sodium octanoate, NaO), a medium chain fatty acid (MCFA), has similar effects. In this study we determined the effect of NaO on S. aureus internalization into bMEC and on the modulation of innate immune elements. NaO (0.25–2 mM) did not affect S. aureus growth and bMEC viability, but it differentially modulated bacterial internalization into bMEC, which was induced at 0.25–0.5 mM (~60%) but inhibited at 1-2 mM (~40%). Also, bMEC showed a basal expression of all the innate immune genes evaluated, which were induced by S. aureus. NaO induced BNBD4, LAP, and BNBD10 mRNA expression, but BNBD5 and TNF-α were inhibited. Additionally, the pretreatment of bMEC with NaO inhibited the mRNA expression induction generated by bacteria which coincides with the increase in internalization; only TAP and BNDB10 showed an increase in their expression; it coincides with the greatest effect on the reduction of bacterial internalization. In conclusion, NaO exerts a dual effect on S. aureus internalization in bMEC and modulates elements of innate immune response.
Collapse
|