Your browser doesn't support javascript.
loading
RNA-Seq identifies condition-specific biological signatures of ischemia-reperfusion injury in the human kidney.
Park, Meeyoung; Kwon, Chae Hwa; Ha, Hong Koo; Han, Miyeun; Song, Sang Heon.
Afiliação
  • Park M; Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea.
  • Kwon CH; Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea.
  • Ha HK; Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea.
  • Han M; Department of Urology, Pusan National University Hospital, Busan, South Korea.
  • Song SH; Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea. shsong0209@gmail.com.
BMC Nephrol ; 21(Suppl 1): 398, 2020 09 25.
Article em En | MEDLINE | ID: mdl-32977749
ABSTRACT

BACKGROUND:

Acute kidney injury (AKI) is defined as a sudden event of kidney failure or kidney damage within a short period. Ischemia-reperfusion injury (IRI) is a critical factor associated with severe AKI and end-stage kidney disease (ESKD). However, the biological mechanisms underlying ischemia and reperfusion are incompletely understood, owing to the complexity of these pathophysiological processes. We aimed to investigate the key biological pathways individually affected by ischemia and reperfusion at the transcriptome level.

RESULTS:

We analyzed the steady-state gene expression pattern of human kidney tissues from normal (pre-ischemia), ischemia, and reperfusion conditions using RNA-sequencing. Conventional differential expression and self-organizing map (SOM) clustering analyses followed by pathway analysis were performed. Differential expression analysis revealed the metabolic pathways dysregulated in ischemia. Cellular assembly, development and migration, and immune response-related pathways were dysregulated in reperfusion. SOM clustering analysis highlighted the ischemia-mediated significant dysregulation in metabolism, apoptosis, and fibrosis-related pathways, while cell growth, migration, and immune response-related pathways were highly dysregulated by reperfusion after ischemia. The expression of pro-apoptotic genes and death receptors was downregulated during ischemia, indicating the existence of a protective mechanism against ischemic injury. Reperfusion induced alterations in the expression of the genes associated with immune response such as inflammasome and antigen representing genes. Further, the genes related to cell growth and migration, such as AKT, KRAS, and those related to Rho signaling, were downregulated, suggestive of injury responses during reperfusion. Semaphorin 4D and plexin B1 levels were also downregulated.

CONCLUSIONS:

We show that specific biological pathways were distinctively involved in ischemia and reperfusion during IRI, indicating that condition-specific therapeutic strategies may be imperative to prevent severe kidney damage after IRI in the clinical setting.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Traumatismo por Reperfusão / Transdução de Sinais / Expressão Gênica / Injúria Renal Aguda / RNA-Seq / Rim Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Revista: BMC Nephrol Assunto da revista: NEFROLOGIA Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Coréia do Sul

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Traumatismo por Reperfusão / Transdução de Sinais / Expressão Gênica / Injúria Renal Aguda / RNA-Seq / Rim Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Revista: BMC Nephrol Assunto da revista: NEFROLOGIA Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Coréia do Sul