Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 699: 149566, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38290176

RESUMO

There is increasing interest in the antimicrobial activity of mannosylerythritol lipids-B (MEL-B) against Gram-positive bacteria such as Staphylococcus aureus (S. aureus). However, the specific molecules involved in MEL-B's antimicrobial action against S. aureus have not been identified. This study utilized the Nebraska transposon mutant library (NTML), which contains 1920 mutants, each lacking three-quarters of the genes found in S. aureus. The NTML was screened to identify mutants resistant to MEL-B. Four mutants (Accession Number: SAUSA300_0904, SAUSA300_0752, SAUSA300_0387, and SAUSA300_2311) largely unaffected by incubation with MEL-B, indicating MEL-B resistance. Despite the strong binding of MEL-B to these mutants, the four molecules encoded by the deleted genes (yjbI, clpP, pbuX, or brpS) in each mutant were not directly recognized by MEL-B. Given that these molecules are not localized on the outer surface of S. aureus and that the antibacterial activity of MEL-B against S. aureus is facilitated by the effective transfer of two antibacterial fatty acids (caprylic acid and myristoleic acid) to S. aureus via ME, the deletion of each of the four molecules may alter the peptidoglycan structure, potentially inhibiting the effective transfer of these antimicrobial fatty acids into S. aureus.


Assuntos
Anti-Infecciosos , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Humanos , Staphylococcus aureus/genética , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/genética , Anti-Infecciosos/farmacologia , Infecções Estafilocócicas/microbiologia , Ácidos Graxos , Testes de Sensibilidade Microbiana
2.
Microbiome ; 10(1): 31, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35184756

RESUMO

BACKGROUND: Establishing fecal microbiota transplantation (FMT) to prevent multifactorial diarrhea in calves is challenging because of the differences in farm management practices, the lack of optimal donors, and recipient selection. In this study, the underlying factors of successful and unsuccessful FMT treatment cases are elucidated, and the potential markers for predicting successful FMT are identified using fecal metagenomics via 16S rRNA gene sequencing, fecal metabolomics via capillary electrophoresis time-of-flight mass spectrometry, and machine learning approaches. RESULTS: Specifically, 20 FMT treatment cases, in which feces from healthy donors were intrarectally transferred into recipient diarrheal calves, were conducted with a success rate of 70%. Selenomonas was identified as a microorganism genus that showed significant donor-recipient compatibility in successful FMT treatments. A strong positive correlation between the microbiome and metabolome data, which is a prerequisite factor for FMT success, was confirmed by Procrustes analysis in successful FMT (r = 0.7439, P = 0.0001). Additionally, weighted gene correlation network analysis confirmed the positively or negatively correlated pairs of bacterial taxa (family Veillonellaceae) and metabolomic features (i.e., amino acids and short-chain fatty acids) responsible for FMT success. Further analysis aimed at establishing criteria for donor selection identified the genus Sporobacter as a potential biomarker in successful donor selection. Low levels of metabolites, such as glycerol 3-phosphate, dihydroxyacetone phosphate, and isoamylamine, in the donor or recipients prior to FMT, are predicted to facilitate FMT. CONCLUSIONS: Overall, we provide the first substantial evidence of the factors related to FMT success or failure; these findings could improve the design of future microbial therapeutics for treating diarrhea in calves. Video abstract.


Assuntos
Diarreia , Transplante de Microbiota Fecal , Animais , Bovinos , Diarreia/microbiologia , Diarreia/terapia , Transplante de Microbiota Fecal/métodos , Fezes/microbiologia , RNA Ribossômico 16S/genética , Resultado do Tratamento
3.
World J Microbiol Biotechnol ; 38(3): 54, 2022 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-35149902

RESUMO

Mannosylerythritol lipid-B (MEL-B), which comprises ester-bonded hydrophilic ME and hydrophobic fatty acids, is a bio-surfactant with various unique properties, including antimicrobial activity against most gram-positive bacteria. The gram-positive Staphylococcus aureus is a causative pathogen of dairy cattle mastitis, which results in considerable economic loss in the dairy industry. Here, we demonstrate the efficacy of MEL-B as a disinfectant against bovine-derived S. aureus and elucidate a mechanism of action of MEL-B in the inhibition of bacterial growth. The growth of bovine mastitis causative S. aureus BM1006 was inhibited when cultured with MEL-B above 10 ppm. The activity of MEL-B required fatty acids (i.e., caprylic and myristoleic acids) as ME, the component of MEL-B lacking fatty acids, did not inhibit the growth of S. aureus even at high concentrations. Importantly, ME-bound fatty acids effectively inhibited the growth of S. aureus when compared with free fatty acids. Specifically, the concentrations of ME-bound fatty acids and free caprylic and myristoleic acids required to inhibit the growth of S. aureus were 10, 1442, and 226 ppm, respectively. The involvement of ME in the antimicrobial activity of MEL-B was confirmed by digestion of MEL-B with alkali, which dissociated ME and fatty acids. These results indicated that a mechanism of action of MEL-B in inhibiting the growth of S. aureus could be explained by the effective transporting of antimicrobial fatty acids to the bacterial surface via hydrophilic ME.


Assuntos
Anti-Infecciosos , Mastite Bovina , Infecções Estafilocócicas , Animais , Antibacterianos/farmacologia , Bovinos , Feminino , Glicolipídeos , Mastite Bovina/tratamento farmacológico , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/veterinária , Staphylococcus aureus
4.
Sci Rep ; 12(1): 931, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35042931

RESUMO

Gland macrophages are primed for gland development and functions through interactions within their niche. However, the phenotype, ontogeny, and function of steady-state salivary gland (SG) macrophages remain unclear. We herein identified CD11c+ and CD11c- subsets among CD64+ macrophages in steady-state murine SGs. CD11c- macrophages were predominant in the SGs of embryonic and newborn mice and decreased with advancing age. CD11c+ macrophages were rarely detected in the embryonic period, but rapidly expanded after birth. CD11c+, but not CD11c-, macrophage numbers decreased in mice treated with a CCR2 antagonist, suggesting that CD11c+ macrophages accumulate from bone marrow-derived progenitors in a CCR2-dependent manner, whereas CD11c- macrophages were derived from embryonic progenitors in SGs. CD11c+ and CD11c- macrophages strongly expressed colony-stimulating factor (CSF)-1 receptor, the injection of an anti-CSF-1 receptor blocking antibody markedly reduced both subsets, and SGs strongly expressed CSF-1, indicating the dependency of SG resident macrophage development on CSF-1. The phagocytic activity of SG macrophages was extremely weak; however, the gene expression profile of SG macrophages indicated that SG macrophages regulate gland development and functions in SGs. These results suggest that SG CD11c+ and CD11c- macrophages are developed and instructed to perform SG-specific functions in steady-state SGs.


Assuntos
Antígenos CD11/genética , Macrófagos/metabolismo , Glândulas Salivares/metabolismo , Animais , Antígenos CD11/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Diferenciação Celular , Células Dendríticas/imunologia , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/imunologia , Masculino , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Fagócitos/metabolismo , Glândulas Salivares/imunologia
5.
Cell Rep ; 36(10): 109655, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34496253

RESUMO

The evolutionary strategy of transferring maternal antibodies via milk profoundly impacts the survival, lifelong health, and wellbeing of all neonates, including a pronounced impact on human breastfeeding success and infant development. While there has been increased recognition that interorgan connectivity influences the quality of a mother's milk, potentially to personalize it for her offspring, the underlying bases for these processes are incompletely resolved. Here, we define an essential role of Peyer's patches (PPs) for the generation of plasma cells that secrete maternal immunoglobulin A (IgA) into milk. Our metagenomic analysis reveals that the presence of certain residential microorganisms in the gastrointestinal (GI) tract, such as Bacteroides acidifaciens and Prevotella buccalis, is indispensable for the programming of maternal IgA synthesis prior to lactational transfer. Our data provide important insights into how the microbiome of the maternal GI environment, specifically through PPs, can be communicated to the next generation via milk.


Assuntos
Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Leite Humano/imunologia , Plasmócitos/citologia , Animais , Humanos , Imunoglobulina A/imunologia , Imunoglobulina A Secretora/imunologia , Camundongos , Nódulos Linfáticos Agregados/imunologia
6.
Cell Mol Gastroenterol Hepatol ; 10(1): 83-100, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32017983

RESUMO

BACKGROUND & AIMS: Organ-level research using an animal model lacking Il2rg, the gene responsible for X-linked severe combined immunodeficiency (X-SCID), is clinically unavailable and would be a powerful tool to gain deeper insights into the symptoms of patients with X-SCID. METHODS: We used an X-SCID animal model, which was first established in our group by the deletion of Il2rg gene in pigs, to understand the clinical signs from multiple perspectives based on pathology, immunology, microbiology, and nutrition. We also treated the X-SCID pigs with bone marrow transplantation (BMT) for mimicking a current therapeutic treatment for patients with X-SCID and investigated the effect at the organ-level. Moreover, the results were confirmed using serum and fecal samples collected from patients with X-SCID. RESULTS: We demonstrated that X-SCID pigs completely lacked Peyer's patches (PPs) and IgA production in the small intestine, but possessed some dysfunctional intestinal T and B cells. Another novel discovery was that X-SCID pigs developed a heterogeneous intestinal microflora and possessed abnormal plasma metabolites, indicating that X-SCID could be an immune disorder that affects various in vivo functions. Importantly, the organogenesis of PPs in X-SCID pigs was not promoted by BMT. Although a few isolated lymphoid follicles developed in the small intestine of BMT-treated X-SCID pigs, there was no evidence that they contributed to IgA production and microflora formation. Consistently, most patients with X-SCID who received BMT possessed abnormal intestinal immune and microbial environments regardless of the presence of sufficient serum IgG. CONCLUSIONS: These results indicate that the current BMT therapies for patients with X-SCID may be insufficient to induce the organogenesis of intestinal lymphoid tissues that are associated with numerous functions in vivo.


Assuntos
Transplante de Medula Óssea , Subunidade gama Comum de Receptores de Interleucina/genética , Mucosa Intestinal/crescimento & desenvolvimento , Nódulos Linfáticos Agregados/crescimento & desenvolvimento , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Adolescente , Adulto , Animais , Animais Geneticamente Modificados , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/imunologia , Técnicas de Inativação de Genes , Humanos , Imunidade nas Mucosas , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Organogênese/genética , Organogênese/imunologia , Nódulos Linfáticos Agregados/imunologia , Suínos , Resultado do Tratamento , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/patologia
7.
Front Immunol ; 11: 604674, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424851

RESUMO

Morphogenesis and differentiation of organs is required for subsequent functional maturation. The morphological features of Peyer's patches vary among species. In pigs, they develop extensively in the ileum as ileal Peyer's patches (IPPs). However, the role of IPPs in the porcine immune system remains to be elucidated because of a lack of complete understanding of IPP organogenesis. Results of the present study revealed that development of porcine IPPs is initiated prenatally between embryonic days 76 and 91. The process of IPP organogenesis is concomitant with increased transcriptional patterns of CXCL13 and CCL19. IPPs undergo further development postnatally by forming central, marginal, and subepithelial zones. Importantly, a large number of proliferating B cells and apoptotic cells are found in porcine IPPs postnatally, but not prenatally. The expression level of IgM in proliferating B cells depends on the zone in which distinct B cells are separately localized after birth. Specifically, IgM+ cells are predominantly found in the central zone, whereas IgM-/low cells are abundant in the marginal zone. Importantly, the cellular feature of IPPs differs from that of mesenteric lymph nodes (MLNs) where such distinct zones are not formed both prenatally and postnatally. Our findings suggest that IPPs (not MLNs) in postnatal pigs are involved in complementing functions of the primary lymphoid tissue that promotes the differentiation and maturation of B cells.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular , Proliferação de Células , Íleo/embriologia , Nódulos Linfáticos Agregados/embriologia , Fatores Etários , Animais , Animais Recém-Nascidos , Apoptose , Linfócitos B/metabolismo , Quimiocina CCL19/genética , Quimiocina CCL19/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Íleo/imunologia , Íleo/metabolismo , Imunoglobulina M/metabolismo , Organogênese , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Gravidez , Sus scrofa , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
8.
Vet Res ; 49(1): 22, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29482613

RESUMO

Staphylococcus aureus is a major pathogen that causes subclinical mastitis associated with huge economic losses to the dairy industry. A few vaccines for bovine mastitis are available, and they are expected to induce the production of S. aureus-specific antibodies that prevent bacterial adherence to host cells or promote opsonization by phagocytes. However, the efficacy of such vaccines are still under debate; therefore, further research focusing on improving the current vaccines by seeking additional mechanisms of action is required to reduce economic losses due to mastitis in the dairy industry. Here, we generated S. aureus-specific bovine IgG antibodies (anti-S. aureus) that directly inhibited bacterial growth in vitro. Inhibition depended on specificity for anti-S. aureus, not the interaction between Protein A and the fragment crystallizable region of the IgG antibodies or bacterial agglutination. An in vitro culture study using S. aureus strain JE2 and its deletion mutant JE2ΔSrtA, which lacks the gene encoding sortase A, revealed that the effect of anti-S. aureus was sortase-A-independent. Sortase A is involved in the synthesis of cell-wall-associated proteins. Thus, other surface molecules, such as membrane proteins, cell surface polysaccharides, or both, may trigger the inhibition of bacterial growth by anti-S. aureus. Together, our findings contribute insights into developing new strategies to further improve the available mastitis vaccine by designing a novel antigen on the surface of S. aureus to induce inhibitory signals that prevent bacterial growth.


Assuntos
Anticorpos Antibacterianos/metabolismo , Doenças dos Bovinos/imunologia , Imunoglobulina G/metabolismo , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/imunologia , Animais , Bovinos , Masculino , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA