Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 851
Filtrar
1.
J Steroid Biochem Mol Biol ; 240: 106508, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38521361

RESUMEN

Mastitis is one the most widespread and serious diseases in dairy cattle. Recurrent and chronic infections are often attributable to certain pathogenicity mechanisms in mastitis-causing pathogens such as Staphylococcus spp. These include growing in biofilm and invading cells, both of which make it possible to resist or evade antimicrobial therapies and the host's immune system. This study tested the effects of active vitamin D3 (i.e., calcitriol or 1,25-dihydroxyvitamin D3) on the internalization and phagocytosis of biofilm-forming Staphylococcus spp. isolated from animals with mastitis. Two established bovine cell lines were used: MAC-T (mammary epithelial cells) and BoMac (macrophages). Calcitriol (0-200 nM) did not affect the viability of MAC-T cells nor that of BoMac cells after 24 and 72 h. Concentrations of 0-100 mM for 24 h upregulated the expression of 24-hydroxylase in MAC-T cells, but did not alter that of VDR. Pre-treatment of the cells with calcitriol for 24 h decreased the internalization of S. aureus V329 into MAC-T cells (0-100 nM), and stimulated the phagocytosis of the same strain and of S. xylosus 4913 (0-10 nM). Calcitriol and two conditioned media, obtained by treating the cells with 25-200 nM of the metabolite for 24 h, were also assessed in terms of their antimicrobial and antibiofilm activity. Neither calcitriol by itself nor the conditioned media affected staphylococcal growth or biofilm formation (0-200 nM for 12 and 24 h, respectively). In contrast, the conditioned media (0-100 nM for 24 h) decreased the biomass of preformed non-aureus staphylococcal biofilms and killed the bacteria within them, without affecting metabolic activity. These effects may be mediated by reactive oxygen species and proteins with antimicrobial and/or antibiofilm activity. In short, calcitriol could make pathogens more accessible to antimicrobial therapies and enhance bacterial clearance by professional phagocytes. Moreover, it may modulate the host's endogenous defenses in the bovine udder and help combat preformed non-aureus staphylococcal biofilms (S. chromogenes 40, S. xylosus 4913, and/or S. haemolyticus 6). The findings confirm calcitriol's potential as an adjuvant to prevent and/or treat intramammary infections caused by Staphylococcus spp., which would in turn contribute to reducing antibiotic use on dairy farms.


Asunto(s)
Biopelículas , Inmunidad Innata , Mastitis Bovina , Fagocitosis , Staphylococcus , Animales , Bovinos , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Femenino , Mastitis Bovina/microbiología , Mastitis Bovina/inmunología , Inmunidad Innata/efectos de los fármacos , Staphylococcus/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Calcitriol/farmacología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/veterinaria , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Línea Celular , Glándulas Mamarias Animales/microbiología , Glándulas Mamarias Animales/inmunología , Macrófagos/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo
2.
Vet Res ; 54(1): 32, 2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37016420

RESUMEN

Host response to invasive microbes in the bovine udder has an important role on the animal health and is essential to the dairy industry to ensure production of high-quality milk and reduce the mastitis incidence. To better understand the biology behind these host-microbiome interactions, we investigated the somatic cell proteomes at quarter level for four cows (collected before and after milking) using a shotgun proteomics approach. Simultaneously, we identified the quarter microbiota by amplicon sequencing to detect presence of mastitis pathogens or other commensal taxa. In total, 32 quarter milk samples were analyzed divided in two groups depending on the somatic cell count (SCC). The high SCC group (>100,000 cell/mL) included 10 samples and significant different proteome profiles were detected. Differential abundance analysis uncovers a specific expression pattern in high SCC samples revealing pathways involved in immune responses such as inflammation, activation of the complement system, migration of immune cells, and tight junctions. Interestingly, different proteome profiles were also identified in quarter samples containing one of the two mastitis pathogens, Staphylococcus aureus and Streptococcus uberis, indicating a different response of the host depending on the pathogen. Weighted correlation network analysis identified three modules of co-expressed proteins which were correlated with the SCC in the quarters. These modules contained proteins assigned to different aspects of the immune response, but also amino sugar and nucleotide sugar metabolism, and biosynthesis of amino acids. The results of this study provide deeper insights on how the proteome expression changes at quarter level in naturally infected cows and pinpoint potential interactions and important biological functions during host-microbe interaction.


Asunto(s)
Interacciones Microbiota-Huesped , Glándulas Mamarias Animales , Leche , Proteoma , Animales , Bovinos , Femenino , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , Recuento de Células/veterinaria , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Mastitis Bovina/inmunología , Mastitis Bovina/microbiología , Leche/citología , Proteoma/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/veterinaria , Interacciones Microbiota-Huesped/inmunología
3.
Microb Pathog ; 171: 105726, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35995255

RESUMEN

Staphylococcus aureus (S. aureus) is a major mastitis-causing pathogen in dairy cows. Dairy cows with mastitis suffer from a decrease in milk yield and protein content. Chlorogenic acid (CGA) is a natural product with anti-inflammatory effects. In this study, we examined the function and mechanism of CGA with regard to its anti-inflammatory effects and evaluated its protective function in milk protein synthesis in bovine mammary epithelial cells (BMECs). BMECs were cultured with and without infection by S. aureus and CGA, and extracellular inflammatory cytokines and amino acids in the medium and milk proteins were determined by ELISA. The function of IL-10RA in anti-inflammatory processes and of SF-1 in milk protein synthesis was assessed by gene silencing. The activity of mTORC1, NF-κB, and STAT5 was examined by western blot. S. aureus caused intracellular infection and upregulated TNF-α, IL-1ß, IL-6, and IL-8, whereas uptake of amino acids and milk protein synthesis were suppressed. CGA mitigated the S. aureus-induced inflammatory response and milk protein synthesis in vitro and in vivo. CGA alleviated S. aureus-induced inhibition of mTORC1 and STAT5 and upregulated IL-10 and IL-10RA. In addition, SF-1 was predicted to be a transcription factor of the milk protein-encoding genes α-LA, ß-LG, and CSN2. S. aureus downregulated SF-1 and CGA reversed the decline in milk protein synthesis due to SF-1 knockdown. Thus, CGA mitigates the inflammatory response that is induced by S. aureus and protects the uptake of amino acids and milk protein synthesis in BMECs.


Asunto(s)
Ácido Clorogénico , Mastitis Bovina , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Antiinflamatorios/farmacología , Bovinos , Ácido Clorogénico/farmacología , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Mastitis Bovina/tratamiento farmacológico , Mastitis Bovina/microbiología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de la Leche/metabolismo , Factor de Transcripción STAT5 , Infecciones Estafilocócicas/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo
4.
Microbiol Spectr ; 10(1): e0251221, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35196821

RESUMEN

Mastitis, a highly prevalent disease in dairy cows, is commonly caused by local infection of the mammary gland. Our previous studies have suggested that the gut microbiota plays an important role in the development of mastitis in mice. However, the effects of rumen microbiota on bovine mastitis and the related mechanisms remain unclear. In this study, we assessed the effects and mechanisms of rumen microbiota on bovine mastitis based on the subacute rumen acidosis (SARA) model induced by feeding Holstein Frisian cows a high-concentrate diet for 8 weeks. Then, the inflammatory responses in the mammary gland and the bacterial communities of rumen fluid, feces, and milk were analyzed. The results showed that SARA induced mastitis symptoms in the mammary gland; activated a systemic inflammatory response; and increased the permeability of the blood-milk barrier, gut barrier, and rumen barrier. Further research showed that lipopolysaccharides (LPS), derived from the gut of SARA cows, translocated into the blood and accumulated in the mammary glands. Furthermore, the abundance of Stenotrophomonas was increased in the rumen of SARA cows, and mastitis was induced by oral administration of Stenotrophomonas in lactating mice. In conclusion, our findings suggested that mastitis is induced by exogenous pathogenic microorganisms as well as by endogenous pathogenic factors. Specifically, the elevated abundance of Stenotrophomonas in the rumen and LPS translocation from the rumen to the mammary gland were important endogenous factors that induced mastitis. Our study provides a foundation for novel therapeutic strategies that target the rumen microbiota in cow mastitis. IMPORTANCE Mastitis is a common and frequently occurring disease of humans and animals, especially in dairy farming, which has caused huge economic losses and brought harmful substance residues, drug-resistant bacteria, and other public health risks. The traditional viewpoint indicates that mastitis is mainly caused by exogenous pathogenic bacteria infecting the mammary gland. Our study found that the occurrence of mastitis was induced by the endogenous pathway. Evidence has shown that rumen-derived LPS enters the mammary gland through blood circulation, damaging the blood-milk barrier and then inducing inflammation of the mammary gland in cows. In addition, a higher abundance of Stenotrophomonas in the rumen was closely associated with the development of mastitis. This study provides a basis for novel therapeutic strategies that exploit the rumen microbiota against mastitis in cows.


Asunto(s)
Microbioma Gastrointestinal , Mastitis Bovina/microbiología , Rumen/microbiología , Animales , Traslocación Bacteriana , Bovinos , Heces/microbiología , Femenino , Lactancia , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Mastitis Bovina/inmunología , Mastitis Bovina/fisiopatología , Leche/metabolismo , Stenotrophomonas/fisiología
5.
Immunology ; 165(2): 206-218, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34775606

RESUMEN

The immune system plays fundamental roles in the mammary gland, shaping developmental processes and controlling inflammation during infection and cancer.Here, we reveal unanticipated heterogeneity in the myeloid cell compartment duringdevelopment of virgin, pregnant, lactating and involuting mouse mammary glands,and in milk. We investigate the functional consequences of individual and compoundchemokine receptor deficiency on cell recruitment. Diverse myeloid cell recruitmentwas also shown in models of sterile inflammation and bacterial infection.Strikingly, we have shown that inflammation and infection can alter the abundanceof terminal end buds, a key developmental structure, within the pubertal mammarygland. This previously unknown effect of inflammatory burden during puberty couldhave important implications for understanding pubertal development.


Asunto(s)
Susceptibilidad a Enfermedades , Mastitis/etiología , Mastitis/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Animales , Biomarcadores , Biopsia , Microambiente Celular/genética , Microambiente Celular/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Granulocitos/inmunología , Granulocitos/metabolismo , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Mastitis/patología , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/patología
6.
Front Immunol ; 12: 770822, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858427

RESUMEN

Cow mastitis, which significantly lowers milk quality, is mainly caused by pathogenic bacteria such as E. coli. Previous studies have suggested that lactic acid bacteria can have antagonistic effects on pathogenic bacteria that cause mastitis. In the current study, we evaluated the in vitro and in vivo alleviative effects of L. plantarum KLDS 1.0344 in mastitis treatment. In vitro antibacterial experiments were performed using bovine mammary epithelial cell (bMEC), followed by in vivo studies involving mastitis mouse models. In vitro results indicate that lactic acid was the primary substance inhibiting the E. coli pathogen. Meanwhile, treatment with L. plantarum KLDS 1.0344 can reduce cytokines' mRNA expression levels in the inflammatory response of bMEC induced by LPS. In vivo, the use of this strain reduced the secretion of inflammatory factors IL-6, IL-1ß, and TNF-α, and decreased the activity of myeloperoxidase (MPO), and inhibited the secretion of p-p65 and p-IκBα. These results indicate that L. plantarum KLDS 1.0344 pretreatment can reduce the expression of inflammatory factors by inhibiting the activation of NF-κB signaling pathway, thus exerting prevent the occurrence of inflammation in vivo. Our findings show that L. plantarum KLDS 1.0344 has excellent properties as an alternative to antibiotics and can be developed into lactic acid bacteria preparation to prevent mastitis disease.


Asunto(s)
Escherichia coli/inmunología , Lactobacillus plantarum/inmunología , Glándulas Mamarias Animales/inmunología , Mastitis/inmunología , Animales , Antibiosis/inmunología , Bovinos , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Escherichia coli/metabolismo , Escherichia coli/fisiología , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Lactobacillus plantarum/fisiología , Lipopolisacáridos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/microbiología , Mastitis/inducido químicamente , Mastitis/microbiología , Ratones Endogámicos BALB C , FN-kappa B/inmunología , FN-kappa B/metabolismo , Transducción de Señal/inmunología
7.
Oxid Med Cell Longev ; 2021: 5048375, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34938382

RESUMEN

Mastitis is mainly induced by gram-negative bacterial infections, causing devastating economic losses to the global cattle industry. Both selenium (Se) and taurine (Tau) exhibit multiple biological effects, including reducing inflammation. However, no studies have reported the protective effect of the combined use of Se and Tau against mastitis, and the underlying mechanisms remain unclear. In this study, lipopolysaccharide (LPS), the vital virulence factor of gram-negative bacteria, was used to construct the in vivo and vitro mastitis models. The results of in vivo model showed that Se and Tau combination was more effective than either substance alone in reducing tissue hyperemia, edema, and neutrophil infiltration in the mammary acinar cavity, improving the blood-milk barrier in LPS-induced mice mastitis, and decreasing the expression of proinflammatory factors and the activity of MPO. Moreover, Se and Tau combination significantly increased the levels of LPS-induced reduction in PI3K/Akt/mTOR, but the expressions of TLRs and NLRP3 were not significantly changed in the mammary tissue. In the in vitro experiments, the effects of Se and Tau combination or alone on inflammatory factors, inflammatory mediators, MPO activity, and blood-milk barrier were consistent with those in vivo. The Se and Tau combination has also been found to increase the survival rate of BMECs compared with each substance alone via promoting cellular proliferation and inhibiting apoptosis. Also, it has been confirmed that this combination could restore the LPS-induced inhibition in the PI3K/Akt/mTOR signaling pathway. Inhibition of mTOR by Rapamycin counteracted the combined protection of SeMet and Tau against LPS-induced inflammatory damage, the inhibition of PI3K by LY294002 blocked the activation of mTOR, and the accumulation of ROS by the ROS agonist blocked the activation of PI3K. In conclusion, these findings suggested that Se and Tau combination was better than either substance alone in protecting LPS-induced mammary inflammatory lesions by upregulating the PI3K/Akt/mTOR signaling pathway.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/prevención & control , Glándulas Mamarias Animales/efectos de los fármacos , Mastitis/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Selenio/farmacología , Taurina/farmacología , Animales , Antiinflamatorios/farmacología , Bovinos , Quimioterapia Combinada , Femenino , Depuradores de Radicales Libres , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Mastitis/inducido químicamente , Mastitis/inmunología , Mastitis/metabolismo , Ratones , Ratones Endogámicos ICR , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
8.
Cells ; 10(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34943933

RESUMEN

Escherichia coli and Staphylococcus aureus are major mastitis causing pathogens in dairy cattle but elicit distinct immune and an inflammatory response in the udder. However, the host determinants responsible for this difference remains largely unknown. Our initial studies focused on the global transcriptomic response of primary bovine mammary epithelial cells (pbMECs) to heat-killed E. coli and S. aureus. RNA-sequencing transcriptome analysis demonstrates a significant difference in expression profiles induced by E. coli compared with S. aureus. A major differential response was the activation of innate immune response by E. coli, but not by S. aureus. Interestingly, E. coli stimulation increased transcript abundance of several genes downstream of Nrf2 (nuclear factor erythroid 2-related factor 2) that were enriched in gene sets with a focus on metabolism and immune system. However, none of these genes was dysregulated by S. aureus. Western blot analysis confirms that S. aureus impairs Nrf2 activation as compared to E. coli. Using Nrf2-knockdown cells we demonstrate that Nrf2 is necessary for bpMECs to mount an effective innate defensive response. In support of this notion, nuclear Nrf2 overexpression augmented S. aureus-stimulated inflammatory response. We also show that, unlike E. coli, S. aureus disrupts the non-canonical p62/SQSTM1-Keap1 pathway responsible for Nrf2 activation through inhibiting p62/SQSTM1 phosphorylation at S349. Collectively, our findings provide important insights into the contribution of the Nrf2 pathway to the pathogen-species specific immune response in bovine mammary epithelial cells and raise a possibility that impairment of Nrf2 activation contributes to, at least in part, the weak inflammatory response in S. aureus mastitis.


Asunto(s)
Inmunidad Innata/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Mastitis/genética , Factor 2 Relacionado con NF-E2/genética , Proteína Sequestosoma-1/genética , Animales , Bovinos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Femenino , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/microbiología , Glándulas Mamarias Animales/patología , Mastitis/inmunología , Mastitis/microbiología , Mastitis/patología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad
9.
Cell Rep ; 37(10): 110099, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879282

RESUMEN

Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/inmunología , Células Epiteliales/inmunología , Activación de Linfocitos , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Células T Asesinas Naturales/inmunología , Paridad , Animales , Antígenos CD1d/metabolismo , Comunicación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Genes BRCA1 , Genes myc , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células T Asesinas Naturales/metabolismo , Embarazo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Transducción de Señal , Microambiente Tumoral
10.
Sci Rep ; 11(1): 22896, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819614

RESUMEN

The determination of the somatic cell count of a milk sample is one of the most common methods to monitor udder health of a dairy cow. However, this procedure does not take into account the fact that cells in milk present a great variety of different cell types. The objective of our study was to establish a high-resolution differential cell count (HRDCC) by means of flow cytometry in blood and milk. We were able to detect ten subpopulations among the three main populations of immune cells and to determine their viability. Additionally, blood samples were analyzed for common laboratory biomarkers, i.e. differential blood counts, haptoglobin levels and several metabolic parameters. In this first feasibility study, we used three different vaccines to stimulate the immune system of five healthy cows each. Samples were collected shortly before, in between and after the vaccinations. Using multivariate statistical methods we saw a diagnostic benefit when HRDCCs were included compared to only the standard laboratory parameters. The impacts of all three vaccinations on the immune system were visible in blood HRDCCs as well as in milk HRDCCs. Cluster of Differentiation 8+ (CD8+) T cells, B cells and monocyte/macrophage subpopulations were among the most important and statistically relevant parameters for all treatments in both biofluids. Moreover, in one of the treatment groups intermediate monocytes showed a significant increase after both vaccinations. Although the use of HRDCC in blood or milk was shown to be highly relevant for early systemic diagnostic, to confirm these subpopulations further investigations in cows of different breed, lactation stage or health status are required.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Industria Lechera , Citometría de Flujo/veterinaria , Inmunofenotipificación/veterinaria , Macrófagos/inmunología , Leche/inmunología , Monocitos/inmunología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Bovinos , Femenino , Macrófagos/efectos de los fármacos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/inmunología , Leche/citología , Monocitos/efectos de los fármacos , Vacunación/veterinaria , Vacunas/administración & dosificación
11.
Front Immunol ; 12: 754661, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745127

RESUMEN

The mammary gland is unique in female mammals. Mammary tissue undergoes development and remodeling during lactation, a stage associated with high susceptibility to bacterial infections, inducing an inflammatory condition called mastitis. Although the immune response of the mammary gland has been the subject of intense research to improve prevention and treatment efficacy, the precise definition of its immune composition at this particular physiological stage is still missing. We combined single-cell RNA-Seq, flow cytometry, and three-dimensional confocal microscopy techniques to characterize the immune landscape of lactating murine mammary tissue. Macrophages dominated the immune cell repertoire and could be subdivided into at least two subsets: ductal and stromal macrophages. Ductal macrophages represented approximately 80% of the total CD45pos immune cells and co-expressed F4/80 and CD11c, with high levels of MHC class II molecules. They were strategically poised below the alveolar basal cells in contact with the myoepithelial cell network. Adaptive T and B lymphocytes were remarkably less numerous at this stage, which could explain the limited efficacy of vaccination against mastitis. These results support the view that new strategies to increase mammary immunity and prevent mastitis should be devised.


Asunto(s)
Lactancia/inmunología , Macrófagos/inmunología , Glándulas Mamarias Animales/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
12.
Int J Mol Sci ; 22(18)2021 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-34575988

RESUMEN

Maedi-Visna-like genotype A strains and Caprine arthritis encephaltis-like genotype B strains are small ruminant lentiviruses (SRLV) which, for incompletely understood reasons, appear to be more virulent in sheep and goats, respectively. A 9-month in vivo infection experiment using Belgian genotype A and B SRLV strains showed that almost all homologous (genotype A in sheep; genotype B in goats) and heterologous (genotype A in goats; genotype B in sheep) intratracheal inoculations resulted in productive infection. No differences in viremia and time to seroconversion were observed between homologous and heterologous infections. Higher viral loads and more severe lesions in the mammary gland and lung were however detected at 9 months post homologous compared to heterologous infection which coincided with strongly increased IFN-γ mRNA expression levels upon homologous infection. Pepscan analysis revealed a strong antibody response against immune-dominant regions of the capsid and surface proteins upon homologous infection, which was absent after heterologous infection. These results inversely correlated with protection against virus replication in target organs and observed histopathological lesions, and thus require an in-depth evaluation of a potential role of antibody dependent enhancement in SRLV infection. Finally, no horizontal intra- and cross-species SRLV transmission to contact animals was detected.


Asunto(s)
Virus de la Artritis-Encefalitis Caprina/fisiología , Genotipo , Enfermedades de las Cabras/inmunología , Cabras , Inmunidad Humoral , Neumonía Intersticial Progresiva de los Ovinos/inmunología , Ovinos , Replicación Viral/inmunología , Virus Visna-Maedi/fisiología , Animales , Anticuerpos Antivirales/inmunología , Femenino , Enfermedades de las Cabras/genética , Enfermedades de las Cabras/patología , Enfermedades de las Cabras/virología , Cabras/inmunología , Cabras/virología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/virología , Neumonía Intersticial Progresiva de los Ovinos/genética , Neumonía Intersticial Progresiva de los Ovinos/patología , Neumonía Intersticial Progresiva de los Ovinos/virología , Ovinos/inmunología , Ovinos/virología , Especificidad de la Especie , Carga Viral/inmunología
13.
Commun Biol ; 4(1): 993, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34417554

RESUMEN

The human breast and ovine mammary gland undergo striking levels of postnatal development, leading to formation of terminal duct lobular units (TDLUs). Here we interrogate aspects of sheep TDLU growth as a model of breast development and to increase understanding of ovine mammogenesis. The distributions of epithelial nuclear Ki67 positivity differ significantly between younger and older lambs. Ki67 expression is polarised to the leading edge of the developing TDLUs. Intraepithelial ductal macrophages exhibit periodicity and considerably increased density in lambs approaching puberty. Stromal macrophages are more abundant centrally than peripherally. Intraepithelial T lymphocytes are more numerous in older lambs. Stromal hotspots of Ki67 expression colocalize with immune cell aggregates that exhibit distinct organisation consistent with tertiary lymphoid structures. The lamb mammary gland thus exhibits a dynamic mucosal and stromal immune microenvironment and constitutes a valuable model system that provides new insights into postnatal breast development.


Asunto(s)
Inmunidad Mucosa/inmunología , Macrófagos/inmunología , Glándulas Mamarias Animales/inmunología , Oveja Doméstica/inmunología , Células del Estroma/inmunología , Animales , Femenino , Macrófagos/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Oveja Doméstica/crecimiento & desarrollo , Células del Estroma/metabolismo
14.
Int Immunopharmacol ; 100: 108079, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34455255

RESUMEN

Mastitis is an inflammation of mammary gland, which directly affects the milk production performance and causes huge economic losses in the dairy industry. During mastitis, the blood-milk barrier (BMB) loses its integrity and aggravates the severity of mastitis. Exogenous DNase I has been exerted protective effects in different model of tissue injury. Here, we designed a study to investigate the effects of DNase I on inflammation and BMB in a mice model of Staphylococcus aureus-induced mastitis. In the model, we found that DNase I treatment significantly alleviated the inflammatory response through decrease of inflammatory cells in mammary alveoli, MPO activity and cytokines in mammary gland. Furthermore, immunofluorescent staining and western blotting demonstrated that exogenous DNase I obviously reduced BMB permeability and changed the expression of tight junction proteins to support the re-establishment of the barrier integrity. Mechanismly, DNase I treatment inhibited NF-κB and enhanced AKT signaling pathways. Therefore, our results indicate that DNase I may be an effective treatment for attenuating mastitis.


Asunto(s)
Desoxirribonucleasa I/farmacología , Mastitis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Desoxirribonucleasa I/uso terapéutico , Femenino , Humanos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Glándulas Mamarias Animales/irrigación sanguínea , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Glándulas Mamarias Animales/patología , Mastitis/inmunología , Mastitis/microbiología , Mastitis/patología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/inmunología
15.
Int J Mol Sci ; 22(12)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200743

RESUMEN

Mastitis is a common disease in dairy cows that is mostly caused by E. coli, and it brings massive losses to the dairy industry. N6-Methyladenosine (m6A), a methylation at the N6 position of RNA adenine, is a type of modification strongly associated with many diseases. However, the role of m6A in mastitis has not been investigated. In this study, we used MeRIP-seq to sequence the RNA of bovine mammary epithelial cells treated with inactivated E. coli for 24 h. In this in vitro infection model, there were 16,691 m6A peaks within 7066 mRNA transcripts in the Con group and 10,029 peaks within 4891 transcripts in the E. coli group. Compared with the Con group, 474 mRNAs were hypermethylated and 2101 mRNAs were hypomethylated in the E. coli group. Biological function analyses revealed differential m6A-modified genes mainly enriched in the MAPK, NF-κB, and TGF-ß signaling pathways. In order to explore the relationship between m6A and mRNA expression, combined MeRIP-seq and mRNA-seq analyses revealed 212 genes with concomitant changes in the mRNA expression and m6A modification. This study is the first to present a map of RNA m6A modification in mastitis treated with E. coli, providing a basis for future research.


Asunto(s)
Adenosina/análogos & derivados , Metilación de ADN , Células Epiteliales/metabolismo , Infecciones por Escherichia coli/veterinaria , Regulación de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Mastitis Bovina/genética , Adenosina/química , Animales , Bovinos , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Femenino , Perfilación de la Expresión Génica , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Mastitis Bovina/inmunología , Mastitis Bovina/microbiología
16.
Front Immunol ; 12: 700278, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34267762

RESUMEN

ß-hydroxybutyrate (BHB) has been associated with disease incidence in early lactation dairy cattle, but such associations do not demonstrate causation. Therefore, the objective of this study was to examine the effects of BHB during an intramammary Streptococcus uberis challenge. A secondary objective was to elucidate the mechanisms behind BHB effects on cytokine transcript abundance using the RAW 264.7 cell line. Late lactation multiparous dairy cows (n = 12) were continuously infused intravenously with either BHB to induce hyperketonemia (target concentration: 1.8 mM) or with saline (CON) for 72 h during a S. uberis intramammary challenge. Body temperature, dry matter intake (DMI), milk production, and milk S. uberis cfu were measured daily until one week post-challenge. Blood samples were collected during infusion to assess changes in metabolism (glucose, insulin, glucagon, NEFA, and cortisol) and systemic inflammation (IL-1ß and SAA). Mammary biopsies were conducted at 72 h post-challenge to assess transcript abundance of inflammation-associated genes. BHB-infused cows exhibited a delayed febrile response, noted by a lesser vaginal temperature during the final day of infusion, followed by a greater vaginal temperature 6 d post-challenge. Consequently, BHB-infused cows had greater S. uberis cfu on d 4, 6, and 7 as compared to CON. Accordingly, BHB-infused cows consumed less DM, produced less milk, had reduced blood glucose, and had increased cortisol concentrations, however, no effects were seen on other systemic parameters or transcript abundance of inflammation-related genes in mammary tissue. To elucidate mechanisms behind the impaired immune defenses, RAW 264.7 cells were transfected with a GPR109A siRNA for 24 h and then treated with or without 1.8 mM BHB and challenged or left unchallenged with S. uberis for an additional 3 h. Transfection with siRNA reduced Gpr109a by 75%. Although BHB treatment did not significantly increase Il10, GPR109A knockdown as compared to the scrambled control reduced Il10 by 90% in S. uberis challenged macrophages treated with BHB, suggesting that macrophage immune responses to S. uberis can be altered via a GPR109A-dependent mechanism. Taken together, these data suggest that BHB altered the immune response promoting tolerance toward S. uberis rather than resistance.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Cetosis/inmunología , Mastitis Bovina/inmunología , Mastitis Bovina/metabolismo , Infecciones Estreptocócicas/metabolismo , Ácido 3-Hidroxibutírico/toxicidad , Animales , Bovinos , Femenino , Cetosis/inducido químicamente , Macrófagos/inmunología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Ratones , Células RAW 264.7 , Infecciones Estreptocócicas/inmunología , Streptococcus
17.
Front Immunol ; 12: 696101, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177964

RESUMEN

Streptococcus uberis (S. uberis) is an important pathogen causing mastitis, which causes continuous inflammation and dysfunction of mammary glands and leads to enormous economic losses. Most research on infection continues to be microbial metabolism-centric, and many overlook the fact that pathogens require energy from host. Mouse is a common animal model for studying bovine mastitis. In this perspective, we uncover metabolic reprogramming during host immune responses is associated with infection-driven inflammation, particularly when caused by intracellular bacteria. Taurine, a metabolic regulator, has been shown to effectively ameliorate metabolic diseases. We evaluated the role of taurine in the metabolic regulation of S. uberis-induced mastitis. Metabolic profiling indicates that S. uberis exposure triggers inflammation and metabolic dysfunction of mammary glands and mammary epithelial cells (the main functional cells in mammary glands). Challenge with S. uberis upregulates glycolysis and oxidative phosphorylation in MECs. Pretreatment with taurine restores metabolic homeostasis, reverses metabolic dysfunction by decrease of lipid, amino acid and especially energy disturbance in the infectious context, and alleviates excessive inflammatory responses. These outcomes depend on taurine-mediated activation of the AMPK-mTOR pathway, which inhibits the over activation of inflammatory responses and alleviates cellular damage. Thus, metabolic homeostasis is essential for reducing inflammation. Metabolic modulation can be used as a prophylactic strategy against mastitis.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Mastitis/prevención & control , Infecciones Estreptocócicas/prevención & control , Streptococcus/patogenicidad , Taurina/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Femenino , Interacciones Huésped-Patógeno , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/microbiología , Mastitis/inmunología , Mastitis/metabolismo , Mastitis/microbiología , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Transducción de Señal , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus/inmunología , Serina-Treonina Quinasas TOR/metabolismo
18.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33958486

RESUMEN

BACKGROUND: Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS: To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS: We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION: Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.


Asunto(s)
Neoplasias de la Mama/inducido químicamente , Transformación Celular Neoplásica/inducido químicamente , Glándulas Mamarias Animales/efectos de los fármacos , Progesterona/administración & dosificación , Receptores de Progesterona/agonistas , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Implantes de Medicamentos , Femenino , Galectina 4/genética , Galectina 4/metabolismo , Inmunidad Innata/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones Transgénicos , Ovariectomía , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transducción de Señal , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
19.
Cell Stem Cell ; 28(10): 1790-1804.e8, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34010627

RESUMEN

The role of heterochromatin in cell fate specification during development is unclear. We demonstrate that loss of the lysine 9 of histone H3 (H3K9) methyltransferase G9a in the mammary epithelium results in de novo chromatin opening, aberrant formation of the mammary ductal tree, impaired stem cell potential, disrupted intraductal polarity, and loss of tissue function. G9a loss derepresses long terminal repeat (LTR) retroviral sequences (predominantly the ERVK family). Transcriptionally activated endogenous retroviruses generate double-stranded DNA (dsDNA) that triggers an antiviral innate immune response, and knockdown of the cytosolic dsDNA sensor Aim2 in G9a knockout (G9acKO) mammary epithelium rescues mammary ductal invasion. Mammary stem cell transplantation into immunocompromised or G9acKO-conditioned hosts shows partial dependence of the G9acKO mammary morphological defects on the inflammatory milieu of the host mammary fat pad. Thus, altering the chromatin accessibility of retroviral elements disrupts mammary gland development and stem cell activity through both cell-autonomous and non-autonomous mechanisms.


Asunto(s)
Retrovirus Endógenos , N-Metiltransferasa de Histona-Lisina , Glándulas Mamarias Animales/crecimiento & desarrollo , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/inmunología , Animales , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Inmunidad , Glándulas Mamarias Animales/inmunología
20.
Cell Rep ; 35(2): 108993, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852863

RESUMEN

Although the contribution of macrophages to metastasis is widely studied in primary tumors, the involvement of macrophages in tumor-draining lymph nodes (LNs) in this process is less clear. We find CD169+ macrophages as the predominant macrophage subtype in naive LNs, which undergo proliferative expansion in response to tumor stimuli. CD169+ LN macrophage depletion, using an anti-CSF-1R antibody or clodronate-loaded liposomes, leads to increased metastatic burden in two mouse breast cancer models. The expansion of CD169+ macrophages is tightly connected to B cell expansion in tumor-draining LNs, and B cell depletion abrogates the effect of CD169+ macrophage absence on metastasis, indicating that the CD169+ macrophage anti-metastatic effects require B cell presence. These results reveal a protective role of CD169+ LN macrophages in breast cancer metastasis and raise caution for the use of drugs aiming at the depletion of tumor-associated macrophages, which might simultaneously deplete macrophages in tumor-draining LNs.


Asunto(s)
Neoplasias Pulmonares/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Biomarcadores/metabolismo , Proliferación Celular , Femenino , Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ganglios Linfáticos/patología , Metástasis Linfática , Macrófagos/citología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , Monocitos/patología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología , Carga Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA