Your browser doesn't support javascript.
loading
Genome-wide DNA hypermethylation opposes healing in patients with chronic wounds by impairing epithelial-mesenchymal transition.
Singh, Kanhaiya; Rustagi, Yashika; Abouhashem, Ahmed S; Tabasum, Saba; Verma, Priyanka; Hernandez, Edward; Pal, Durba; Khona, Dolly K; Mohanty, Sujit K; Kumar, Manishekhar; Srivastava, Rajneesh; Guda, Poornachander R; Verma, Sumit S; Mahajan, Sanskruti; Killian, Jackson A; Walker, Logan A; Ghatak, Subhadip; Mathew-Steiner, Shomita S; Wanczyk, Kristen E; Liu, Sheng; Wan, Jun; Yan, Pearlly; Bundschuh, Ralf; Khanna, Savita; Gordillo, Gayle M; Murphy, Michael P; Roy, Sashwati; Sen, Chandan K.
Afiliação
  • Singh K; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Rustagi Y; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Abouhashem AS; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Tabasum S; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Verma P; Sharkia Clinical Research Department, Ministry of Health, Zagazig, Sharkia, Egypt.
  • Hernandez E; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Pal D; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Khona DK; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Mohanty SK; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Kumar M; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Srivastava R; Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, India.
  • Guda PR; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Verma SS; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Mahajan S; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Killian JA; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Walker LA; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Ghatak S; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Mathew-Steiner SS; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Wanczyk KE; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Liu S; Department of Physics, The Ohio State University, Columbus, Ohio, USA.
  • Wan J; Department of Physics, The Ohio State University, Columbus, Ohio, USA.
  • Yan P; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Bundschuh R; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Khanna S; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Gordillo GM; Department of Surgery, The Ohio State University, Columbus, Ohio, USA.
  • Murphy MP; Indiana University Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Roy S; Center for Computational Biology and Bioinformatics (CCBB), Indiana University School of Medicine, Indianapolis, Indiana, USA.
  • Sen CK; Center for Computational Biology and Bioinformatics (CCBB), Indiana University School of Medicine, Indianapolis, Indiana, USA.
J Clin Invest ; 132(17)2022 09 01.
Article em En | MEDLINE | ID: mdl-35819852
ABSTRACT
An extreme chronic wound tissue microenvironment causes epigenetic gene silencing. An unbiased whole-genome methylome was studied in the wound-edge tissue of patients with chronic wounds. A total of 4,689 differentially methylated regions (DMRs) were identified in chronic wound-edge skin compared with unwounded human skin. Hypermethylation was more frequently observed (3,661 DMRs) in the chronic wound-edge tissue compared with hypomethylation (1,028 DMRs). Twenty-six hypermethylated DMRs were involved in epithelial-mesenchymal transition (EMT). Bisulfite sequencing validated hypermethylation of a predicted specific upstream regulator TP53. RNA-Seq analysis was performed to qualify findings from methylome analysis. Analysis of the downregulated genes identified the TP53 signaling pathway as being significantly silenced. Direct comparison of hypermethylation and downregulated genes identified 4 genes, ADAM17, NOTCH, TWIST1, and SMURF1, that functionally represent the EMT pathway. Single-cell RNA-Seq studies revealed that these effects on gene expression were limited to the keratinocyte cell compartment. Experimental murine studies established that tissue ischemia potently induces wound-edge gene methylation and that 5'-azacytidine, inhibitor of methylation, improved wound closure. To specifically address the significance of TP53 methylation, keratinocyte-specific editing of TP53 methylation at the wound edge was achieved by a tissue nanotransfection-based CRISPR/dCas9 approach. This work identified that reversal of methylation-dependent keratinocyte gene silencing represents a productive therapeutic strategy to improve wound closure.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Metilação de DNA / Transição Epitelial-Mesenquimal Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Metilação de DNA / Transição Epitelial-Mesenquimal Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article