File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Minocycline modulates NFκB phosphorylation and enhances antimicrobial activity against Staphylococcus aureus in mesenchymal stromal/stem cells

TitleMinocycline modulates NFκB phosphorylation and enhances antimicrobial activity against Staphylococcus aureus in mesenchymal stromal/stem cells
Authors
KeywordsMesenchymal stromal cells
Mesenchymal stem cells
Biomaterials
Minocycline
NFκB
Issue Date2017
PublisherBioMed Central Ltd. The Journal's web site is located at http://www.stemcellres.com
Citation
Stem Cell Research & Therapy, 2017, v. 8 n. 1, p. article no. 171 How to Cite?
AbstractBackground: Mesenchymal stromal/stem cells (MSCs) have demonstrated pro-healing properties due to their anti-inflammatory, angiogenic, and even antibacterial properties. We have shown previously that minocycline enhances the wound healing phenotype of MSCs, and MSCs encapsulated in poly(ethylene glycol) and gelatin-based hydrogels with minocycline have antibacterial properties against Staphylococcus aureus (SA). Here, we investigated the signaling pathway that minocycline modulates in MSCs which results in their enhanced wound healing phenotype and determined whether preconditioning MSCs with minocycline has an effect on antimicrobial activity. We further investigated the in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels in inoculated full-thickness cutaneous wounds. Methods: Modulation of cell signaling pathways in MSCs with minocycline was analyzed via western blot, immunofluorescence, and ELISA. Antimicrobial efficacy of MSCs pretreated with minocycline was determined by direct and transwell coculture with SA. MSC viability after SA coculture was determined via a LIVE/DEAD® stain. Internalization of SA by MSCs pretreated with minocycline was determined via confocal imaging. All protein and cytokine analysis was done via ELISA. The in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels was determined in Sprague–Dawley rats inoculated with SA. Two-way ANOVA for multiple comparisons was used with Bonferroni test assessment and an unpaired two-tailed Student’s t test was used to determine p values for all assays with multiple or two conditions, respectively. Results: Minocycline leads to the phosphorylation of transcriptional nuclear factor-κB (NFκB), but not c-Jun NH2-terminal kinase (JNK) or mitogen-activated protein kinase (ERK). Inhibition of NFκB activation prevented the minocycline-induced increase in VEGF secretion. Preconditioning of MSCs with minocycline led to a reduced production of the antimicrobial peptide LL-37, but enhanced antimicrobial activity against SA via an increased production of IL-6 and SA internalization. MSC and antibiotic-loaded hydrogels reduced SA bioburden in inoculated wounds over 3 days and accelerated reepithelialization. Conclusions: Minocycline modulates the NFκB pathway in MSCs that leads to an enhanced production of IL-6 and internalization of SA. This mechanism may have contributed to the in-vivo antibacterial efficacy of MSC and antibiotic-loaded hydrogels.
Persistent Identifierhttp://hdl.handle.net/10722/287180
ISSN
2021 Impact Factor: 8.079
2020 SCImago Journal Rankings: 1.599
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorGuerra, AD-
dc.contributor.authorRose, WE-
dc.contributor.authorHematti, P-
dc.contributor.authorKao, WJ-
dc.date.accessioned2020-09-22T02:57:02Z-
dc.date.available2020-09-22T02:57:02Z-
dc.date.issued2017-
dc.identifier.citationStem Cell Research & Therapy, 2017, v. 8 n. 1, p. article no. 171-
dc.identifier.issn1757-6512-
dc.identifier.urihttp://hdl.handle.net/10722/287180-
dc.description.abstractBackground: Mesenchymal stromal/stem cells (MSCs) have demonstrated pro-healing properties due to their anti-inflammatory, angiogenic, and even antibacterial properties. We have shown previously that minocycline enhances the wound healing phenotype of MSCs, and MSCs encapsulated in poly(ethylene glycol) and gelatin-based hydrogels with minocycline have antibacterial properties against Staphylococcus aureus (SA). Here, we investigated the signaling pathway that minocycline modulates in MSCs which results in their enhanced wound healing phenotype and determined whether preconditioning MSCs with minocycline has an effect on antimicrobial activity. We further investigated the in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels in inoculated full-thickness cutaneous wounds. Methods: Modulation of cell signaling pathways in MSCs with minocycline was analyzed via western blot, immunofluorescence, and ELISA. Antimicrobial efficacy of MSCs pretreated with minocycline was determined by direct and transwell coculture with SA. MSC viability after SA coculture was determined via a LIVE/DEAD® stain. Internalization of SA by MSCs pretreated with minocycline was determined via confocal imaging. All protein and cytokine analysis was done via ELISA. The in-vivo antimicrobial efficacy of MSC and antibiotic-loaded hydrogels was determined in Sprague–Dawley rats inoculated with SA. Two-way ANOVA for multiple comparisons was used with Bonferroni test assessment and an unpaired two-tailed Student’s t test was used to determine p values for all assays with multiple or two conditions, respectively. Results: Minocycline leads to the phosphorylation of transcriptional nuclear factor-κB (NFκB), but not c-Jun NH2-terminal kinase (JNK) or mitogen-activated protein kinase (ERK). Inhibition of NFκB activation prevented the minocycline-induced increase in VEGF secretion. Preconditioning of MSCs with minocycline led to a reduced production of the antimicrobial peptide LL-37, but enhanced antimicrobial activity against SA via an increased production of IL-6 and SA internalization. MSC and antibiotic-loaded hydrogels reduced SA bioburden in inoculated wounds over 3 days and accelerated reepithelialization. Conclusions: Minocycline modulates the NFκB pathway in MSCs that leads to an enhanced production of IL-6 and internalization of SA. This mechanism may have contributed to the in-vivo antibacterial efficacy of MSC and antibiotic-loaded hydrogels.-
dc.languageeng-
dc.publisherBioMed Central Ltd. The Journal's web site is located at http://www.stemcellres.com-
dc.relation.ispartofStem Cell Research & Therapy-
dc.rightsStem Cell Research & Therapy. Copyright © BioMed Central Ltd.-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectMesenchymal stromal cells-
dc.subjectMesenchymal stem cells-
dc.subjectBiomaterials-
dc.subjectMinocycline-
dc.subjectNFκB-
dc.titleMinocycline modulates NFκB phosphorylation and enhances antimicrobial activity against Staphylococcus aureus in mesenchymal stromal/stem cells-
dc.typeArticle-
dc.identifier.emailKao, WJ: wjkao@hku.hk-
dc.identifier.authorityKao, WJ=rp02076-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1186/s13287-017-0623-1-
dc.identifier.pmid28732530-
dc.identifier.pmcidPMC5521110-
dc.identifier.scopuseid_2-s2.0-85025160429-
dc.identifier.hkuros314501-
dc.identifier.volume8-
dc.identifier.issue1-
dc.identifier.spagearticle no. 171-
dc.identifier.epagearticle no. 171-
dc.identifier.isiWOS:000406579700001-
dc.publisher.placeUnited Kingdom-
dc.identifier.issnl1757-6512-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats