Cargando…

Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients

The role of the host in development of persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is not well understood. A cohort of prospectively enrolled patients with persistent methicillin-resistant S. aureus bacteremia (PB) and resolving methicillin-resistant S. aureus bacteremia...

Descripción completa

Detalles Bibliográficos
Autores principales: Mba Medie, Felix, Sharma-Kuinkel, Batu K., Ruffin, Felicia, Chan, Liana C., Rossetti, Maura, Chang, Yu-Ling, Park, Lawrence P., Bayer, Arnold S., Filler, Scott G., Ahn, Richard, Reed, Elaine F., Gjertson, David, Yeaman, Michael R., Fowler, Vance G.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: National Academy of Sciences 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6778225/
https://www.ncbi.nlm.nih.gov/pubmed/31527248
http://dx.doi.org/10.1073/pnas.1909849116
_version_ 1783456732629958656
author Mba Medie, Felix
Sharma-Kuinkel, Batu K.
Ruffin, Felicia
Chan, Liana C.
Rossetti, Maura
Chang, Yu-Ling
Park, Lawrence P.
Bayer, Arnold S.
Filler, Scott G.
Ahn, Richard
Reed, Elaine F.
Gjertson, David
Yeaman, Michael R.
Fowler, Vance G.
author_facet Mba Medie, Felix
Sharma-Kuinkel, Batu K.
Ruffin, Felicia
Chan, Liana C.
Rossetti, Maura
Chang, Yu-Ling
Park, Lawrence P.
Bayer, Arnold S.
Filler, Scott G.
Ahn, Richard
Reed, Elaine F.
Gjertson, David
Yeaman, Michael R.
Fowler, Vance G.
author_sort Mba Medie, Felix
collection PubMed
description The role of the host in development of persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is not well understood. A cohort of prospectively enrolled patients with persistent methicillin-resistant S. aureus bacteremia (PB) and resolving methicillin-resistant S. aureus bacteremia (RB) matched by sex, age, race, hemodialysis status, diabetes mellitus, and presence of implantable medical device was studied to gain insights into this question. One heterozygous g.25498283A > C polymorphism located in the DNMT3A intronic region of chromosome 2p with no impact in messenger RNA (mRNA) expression was more common in RB (21 of 34, 61.8%) than PB (3 of 34, 8.8%) patients (P = 7.8 × 10(−6)). Patients with MRSA bacteremia and g.25498283A > C genotype exhibited significantly higher levels of methylation in gene-regulatory CpG island regions (Δmethylation = 4.1%, P < 0.0001) and significantly lower serum levels of interleukin-10 (IL-10) than patients with MRSA bacteremia without DNMT3A mutation (A/C: 9.7038 pg/mL vs. A/A: 52.9898 pg/mL; P = 0.0042). Expression of DNMT3A was significantly suppressed in patients with S. aureus bacteremia and in S. aureus-challenged primary human macrophages. Small interfering RNA (siRNA) silencing of DNMT3A expression in human macrophages caused increased IL-10 response upon S. aureus stimulation. Treating macrophages with methylation inhibitor 5-Aza-2′-deoxycytidine resulted in increased levels of IL-10 when challenged with S. aureus. In the murine sepsis model, methylation inhibition increased susceptibility to S. aureus. These findings indicate that g.25498283A > C genotype within DNMT3A contributes to increased capacity to resolve MRSA bacteremia, potentially through a mechanism involving increased methylation of gene-regulatory regions and reduced levels of antiinflammatory cytokine IL-10.
format Online
Article
Text
id pubmed-6778225
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher National Academy of Sciences
record_format MEDLINE/PubMed
spelling pubmed-67782252019-10-09 Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients Mba Medie, Felix Sharma-Kuinkel, Batu K. Ruffin, Felicia Chan, Liana C. Rossetti, Maura Chang, Yu-Ling Park, Lawrence P. Bayer, Arnold S. Filler, Scott G. Ahn, Richard Reed, Elaine F. Gjertson, David Yeaman, Michael R. Fowler, Vance G. Proc Natl Acad Sci U S A PNAS Plus The role of the host in development of persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is not well understood. A cohort of prospectively enrolled patients with persistent methicillin-resistant S. aureus bacteremia (PB) and resolving methicillin-resistant S. aureus bacteremia (RB) matched by sex, age, race, hemodialysis status, diabetes mellitus, and presence of implantable medical device was studied to gain insights into this question. One heterozygous g.25498283A > C polymorphism located in the DNMT3A intronic region of chromosome 2p with no impact in messenger RNA (mRNA) expression was more common in RB (21 of 34, 61.8%) than PB (3 of 34, 8.8%) patients (P = 7.8 × 10(−6)). Patients with MRSA bacteremia and g.25498283A > C genotype exhibited significantly higher levels of methylation in gene-regulatory CpG island regions (Δmethylation = 4.1%, P < 0.0001) and significantly lower serum levels of interleukin-10 (IL-10) than patients with MRSA bacteremia without DNMT3A mutation (A/C: 9.7038 pg/mL vs. A/A: 52.9898 pg/mL; P = 0.0042). Expression of DNMT3A was significantly suppressed in patients with S. aureus bacteremia and in S. aureus-challenged primary human macrophages. Small interfering RNA (siRNA) silencing of DNMT3A expression in human macrophages caused increased IL-10 response upon S. aureus stimulation. Treating macrophages with methylation inhibitor 5-Aza-2′-deoxycytidine resulted in increased levels of IL-10 when challenged with S. aureus. In the murine sepsis model, methylation inhibition increased susceptibility to S. aureus. These findings indicate that g.25498283A > C genotype within DNMT3A contributes to increased capacity to resolve MRSA bacteremia, potentially through a mechanism involving increased methylation of gene-regulatory regions and reduced levels of antiinflammatory cytokine IL-10. National Academy of Sciences 2019-10-01 2019-09-16 /pmc/articles/PMC6778225/ /pubmed/31527248 http://dx.doi.org/10.1073/pnas.1909849116 Text en Copyright © 2019 the Author(s). Published by PNAS. https://creativecommons.org/licenses/by-nc-nd/4.0/ https://creativecommons.org/licenses/by-nc-nd/4.0/This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND) (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle PNAS Plus
Mba Medie, Felix
Sharma-Kuinkel, Batu K.
Ruffin, Felicia
Chan, Liana C.
Rossetti, Maura
Chang, Yu-Ling
Park, Lawrence P.
Bayer, Arnold S.
Filler, Scott G.
Ahn, Richard
Reed, Elaine F.
Gjertson, David
Yeaman, Michael R.
Fowler, Vance G.
Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title_full Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title_fullStr Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title_full_unstemmed Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title_short Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients
title_sort genetic variation of dna methyltransferase-3a contributes to protection against persistent mrsa bacteremia in patients
topic PNAS Plus
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6778225/
https://www.ncbi.nlm.nih.gov/pubmed/31527248
http://dx.doi.org/10.1073/pnas.1909849116
work_keys_str_mv AT mbamediefelix geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT sharmakuinkelbatuk geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT ruffinfelicia geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT chanlianac geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT rossettimaura geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT changyuling geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT parklawrencep geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT bayerarnolds geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT fillerscottg geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT ahnrichard geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT reedelainef geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT gjertsondavid geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT yeamanmichaelr geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT fowlervanceg geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients
AT geneticvariationofdnamethyltransferase3acontributestoprotectionagainstpersistentmrsabacteremiainpatients