TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes

Detalhes bibliográficos
Autor(a) principal: Huang,Yayi
Data de Publicação: 2019
Outros Autores: Zhou,Fang, Xiao,Yeda, Shen,Cheng, Liu,Kang, Zhao,Bo
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Revista da Associação Médica Brasileira (Online)
Texto Completo: http://old.scielo.br/scielo.php?script=sci_arttext&pid=S0104-42302019000801067
Resumo: SUMMARY OBJECTIVE Diabetes is a risk factor for acute kidney injury (AKI). However, its mechanism of pathogenesis has not been elucidated. The aim of the study was to investigate the role of inflammation and the toll-like receptor 7 (TLR7) in ischemic AKI for diabetes. METHODS A high glucose hypoxia-reoxygenation model of human renal tubular epithelial (HK-2) cells was used to generate AKI induced by ischemia-reperfusion in diabetes. The activity of cells was measured by CCK-8 assay and LDH activity. Inflammatory cytokines were assessed by ELISA. TLR7, MyD88, and NF-κB expressions were examined by western blotting. Apoptosis was evaluated by flow cytometry. RESULTS The high glucose group and low glucose group were subjected to hypoxia-reoxygenation. The low glucose group developed only mild cell damage, apoptosis, and inflammatory response. In contrast, an equivalent hypoxia-reoxygenation injury provoked severe cell damage, apoptosis, and inflammatory response in the high glucose group. Expression of TLR7 and its related proteins were measured in the high glucose group before and after hypoxia-reoxygenation. The high glucose group exhibited more significant increases in TLR7 expression following hypoxia-reoxygenation than the low glucose group. In addition, the expression of TLR7 and its related proteins after hypoxia-reoxygenation were higher in the high glucose group than in the low glucose group. Inhibition of TLR7 provides significant protection against ischemic injury in diabetes. CONCLUSION Our results suggest that diabetes increases the vulnerability to ischemia-induced renal injury. This increased vulnerability originates from a heightened inflammatory response involving the TLR7 signal transduction pathway.
id AMB-1_4539a3df3cf96a162ebee88a2fa9fdc3
oai_identifier_str oai:scielo:S0104-42302019000801067
network_acronym_str AMB-1
network_name_str Revista da Associação Médica Brasileira (Online)
repository_id_str
spelling TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetesHealth VulnerabilityIschemiaAcute kidney injuryDiabetes mellitusToll-Like Receptor 7SUMMARY OBJECTIVE Diabetes is a risk factor for acute kidney injury (AKI). However, its mechanism of pathogenesis has not been elucidated. The aim of the study was to investigate the role of inflammation and the toll-like receptor 7 (TLR7) in ischemic AKI for diabetes. METHODS A high glucose hypoxia-reoxygenation model of human renal tubular epithelial (HK-2) cells was used to generate AKI induced by ischemia-reperfusion in diabetes. The activity of cells was measured by CCK-8 assay and LDH activity. Inflammatory cytokines were assessed by ELISA. TLR7, MyD88, and NF-κB expressions were examined by western blotting. Apoptosis was evaluated by flow cytometry. RESULTS The high glucose group and low glucose group were subjected to hypoxia-reoxygenation. The low glucose group developed only mild cell damage, apoptosis, and inflammatory response. In contrast, an equivalent hypoxia-reoxygenation injury provoked severe cell damage, apoptosis, and inflammatory response in the high glucose group. Expression of TLR7 and its related proteins were measured in the high glucose group before and after hypoxia-reoxygenation. The high glucose group exhibited more significant increases in TLR7 expression following hypoxia-reoxygenation than the low glucose group. In addition, the expression of TLR7 and its related proteins after hypoxia-reoxygenation were higher in the high glucose group than in the low glucose group. Inhibition of TLR7 provides significant protection against ischemic injury in diabetes. CONCLUSION Our results suggest that diabetes increases the vulnerability to ischemia-induced renal injury. This increased vulnerability originates from a heightened inflammatory response involving the TLR7 signal transduction pathway.Associação Médica Brasileira2019-08-01info:eu-repo/semantics/articleinfo:eu-repo/semantics/publishedVersiontext/htmlhttp://old.scielo.br/scielo.php?script=sci_arttext&pid=S0104-42302019000801067Revista da Associação Médica Brasileira v.65 n.8 2019reponame:Revista da Associação Médica Brasileira (Online)instname:Associação Médica Brasileira (AMB)instacron:AMB10.1590/1806-9282.65.8.1067info:eu-repo/semantics/openAccessHuang,YayiZhou,FangXiao,YedaShen,ChengLiu,KangZhao,Boeng2020-01-20T00:00:00Zoai:scielo:S0104-42302019000801067Revistahttps://ramb.amb.org.br/ultimas-edicoes/#https://old.scielo.br/oai/scielo-oai.php||ramb@amb.org.br1806-92820104-4230opendoar:2020-01-20T00:00Revista da Associação Médica Brasileira (Online) - Associação Médica Brasileira (AMB)false
dc.title.none.fl_str_mv TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
title TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
spellingShingle TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
Huang,Yayi
Health Vulnerability
Ischemia
Acute kidney injury
Diabetes mellitus
Toll-Like Receptor 7
title_short TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
title_full TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
title_fullStr TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
title_full_unstemmed TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
title_sort TLR7 mediates increased vulnerability to ischemic acute kidney injury in diabetes
author Huang,Yayi
author_facet Huang,Yayi
Zhou,Fang
Xiao,Yeda
Shen,Cheng
Liu,Kang
Zhao,Bo
author_role author
author2 Zhou,Fang
Xiao,Yeda
Shen,Cheng
Liu,Kang
Zhao,Bo
author2_role author
author
author
author
author
dc.contributor.author.fl_str_mv Huang,Yayi
Zhou,Fang
Xiao,Yeda
Shen,Cheng
Liu,Kang
Zhao,Bo
dc.subject.por.fl_str_mv Health Vulnerability
Ischemia
Acute kidney injury
Diabetes mellitus
Toll-Like Receptor 7
topic Health Vulnerability
Ischemia
Acute kidney injury
Diabetes mellitus
Toll-Like Receptor 7
description SUMMARY OBJECTIVE Diabetes is a risk factor for acute kidney injury (AKI). However, its mechanism of pathogenesis has not been elucidated. The aim of the study was to investigate the role of inflammation and the toll-like receptor 7 (TLR7) in ischemic AKI for diabetes. METHODS A high glucose hypoxia-reoxygenation model of human renal tubular epithelial (HK-2) cells was used to generate AKI induced by ischemia-reperfusion in diabetes. The activity of cells was measured by CCK-8 assay and LDH activity. Inflammatory cytokines were assessed by ELISA. TLR7, MyD88, and NF-κB expressions were examined by western blotting. Apoptosis was evaluated by flow cytometry. RESULTS The high glucose group and low glucose group were subjected to hypoxia-reoxygenation. The low glucose group developed only mild cell damage, apoptosis, and inflammatory response. In contrast, an equivalent hypoxia-reoxygenation injury provoked severe cell damage, apoptosis, and inflammatory response in the high glucose group. Expression of TLR7 and its related proteins were measured in the high glucose group before and after hypoxia-reoxygenation. The high glucose group exhibited more significant increases in TLR7 expression following hypoxia-reoxygenation than the low glucose group. In addition, the expression of TLR7 and its related proteins after hypoxia-reoxygenation were higher in the high glucose group than in the low glucose group. Inhibition of TLR7 provides significant protection against ischemic injury in diabetes. CONCLUSION Our results suggest that diabetes increases the vulnerability to ischemia-induced renal injury. This increased vulnerability originates from a heightened inflammatory response involving the TLR7 signal transduction pathway.
publishDate 2019
dc.date.none.fl_str_mv 2019-08-01
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://old.scielo.br/scielo.php?script=sci_arttext&pid=S0104-42302019000801067
url http://old.scielo.br/scielo.php?script=sci_arttext&pid=S0104-42302019000801067
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv 10.1590/1806-9282.65.8.1067
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv text/html
dc.publisher.none.fl_str_mv Associação Médica Brasileira
publisher.none.fl_str_mv Associação Médica Brasileira
dc.source.none.fl_str_mv Revista da Associação Médica Brasileira v.65 n.8 2019
reponame:Revista da Associação Médica Brasileira (Online)
instname:Associação Médica Brasileira (AMB)
instacron:AMB
instname_str Associação Médica Brasileira (AMB)
instacron_str AMB
institution AMB
reponame_str Revista da Associação Médica Brasileira (Online)
collection Revista da Associação Médica Brasileira (Online)
repository.name.fl_str_mv Revista da Associação Médica Brasileira (Online) - Associação Médica Brasileira (AMB)
repository.mail.fl_str_mv ||ramb@amb.org.br
_version_ 1754212834294628352