Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Org Lett ; 26(15): 3158-3163, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38588324

ABSTRACT

We uncovered the biosynthetic pathway of the lethal mycotoxin 3-nitropropanoic acid (3-NPA) from koji mold Aspergillus oryzae. The biosynthetic gene cluster (BGC) of 3-NPA, which encodes an amine oxidase and a decarboxylase, is conserved in many fungi used in food processing, although most of the strains have not been reported to produce 3-NPA. Our discovery will lead to efforts that improve the safety profiles of these indispensable microorganisms in making food, alcoholic beverages, and seasoning.


Subject(s)
Aspergillus oryzae , Mycotoxins , Mycotoxins/metabolism , Nitro Compounds , Propionates , Aspergillus oryzae/genetics , Aspergillus oryzae/metabolism
2.
Proc Natl Acad Sci U S A ; 115(47): E11111-E11119, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30297395

ABSTRACT

Staphylococcus aureus is the leading cause of skin and skin structure infection (SSSI), a primary portal of entry for invasive infection. Our prior studies discovered a role for protective innate memory against recurrent methicillin-resistant S. aureus (MRSA) SSSI. In the present study, the dynamics and mechanisms of this response were explored in recurrent SSSI in WT mice. Priming by prior infection reduced skin lesion severity and MRSA burden, and protected against dissemination at day 7 but not day 2. Cytokine and cellular signatures in SSSI differed at day 2 versus 7, and were distinct in skin versus blood or spleen. Cytokines associated with protection in skin included increased IL-17, IL-6, monokine inducible by IFN-γ (MIG), and RANTES, while increased IP-10 correlated with protection from dissemination. Cellular signatures of protection included increased Th17, M1 macrophage, and dendritic cell populations in abscesses, and total macrophages in lymph nodes. Priming potentiated S. aureus-specific phagocytic killing by bone marrow-derived macrophages in vitro, and their adoptive transfer into naïve skin afforded protective efficacy in vivo. Present findings indicate that protective immunity in recurrent S. aureus infection is locally targeted, and involves specific memory conferred by macrophages. These insights provide targets for vaccine and immunotherapeutic development against MRSA.


Subject(s)
Immunity, Innate/immunology , Immunologic Memory/immunology , Macrophages/immunology , Macrophages/transplantation , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Skin Infections/immunology , Adoptive Transfer , Animals , Chemokine CCL5/blood , Chemokine CXCL10/blood , Dendritic Cells/immunology , Disease Models, Animal , Homeodomain Proteins/genetics , Interferon-gamma/blood , Interleukin-17/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Skin Infections/microbiology , Th17 Cells/immunology
3.
Infect Immun ; 85(2)2017 02.
Article in English | MEDLINE | ID: mdl-27849182

ABSTRACT

Staphylococcus aureus is the leading cause of skin and skin structure infections (SSSI). The high frequency of recurring SSSI due to S. aureus, including methicillin-resistant S. aureus (MRSA) strains, despite high titers of specific antibodies and circulating T cells, implies that traditional adaptive immunity imparts incomplete protection. We hypothesized that innate immune memory contributes to the protective host defense against recurring MRSA infection. To test this hypothesis, SSSI was induced in wild-type and rag1-/- mice in the BALB/c and C57BL/6 backgrounds. Prior infection (priming) of wild-type and rag1-/- mice of either background afforded protection against repeat infection, as evidenced by reduced abscess severities and decreased CFU densities compared to those in naive controls. Interestingly, protection was greater on the previously infected flank than on the naive flank for wild-type and rag1-/- mice. For wild-type mice, protective efficacy corresponded to increased infiltration of neutrophils (polymorphonuclear leukocytes [PMN]), macrophages (MΦ), Langerin+ dendritic cells (LDC), and natural killer (NK) cells. Protection was associated with the induction of interleukin-17A (IL-17A), IL-22, and gamma interferon (IFN-γ) as well as the antimicrobial peptides CRAMP and mßD-3. Priming also protected rag1-/- mice against recurring SSSI, with increased MΦ and LDC infiltration and induction of IL-22, CRAMP, and mßD-3. These findings suggest that innate immune memory, mediated by specific cellular and molecular programs, likely contributes to the localized host defense in recurrent MRSA SSSI. These insights support the development of targeted immunotherapeutic strategies to address the challenge of MRSA infection.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate , Immunologic Memory , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression , Male , Mice , Mice, Knockout , Recurrence , Spleen/cytology , Spleen/immunology , Staphylococcal Skin Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...